+ All Categories
Home > Documents > Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of...

Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of...

Date post: 05-Oct-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
88
Comprehensive Summaries of Uppsala Dissertations from the Faculty of Pharmacy 263 Computational Studies of HIV-1 Protease Inhibitors BY WESLEY SCHAAL ACTA UNIVERSITATIS UPSALIENSIS UPPSALA 2002
Transcript
Page 1: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

Comprehensive Summaries of Uppsala Dissertationsfrom the Faculty of Pharmacy 263

Computational Studies of HIV-1Protease Inhibitors

BY

WESLEY SCHAAL

ACTA UNIVERSITATIS UPSALIENSISUPPSALA 2002

Page 2: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

Dissertation for the Degree of Doctor of Philosophy (Faculty of Pharmacy) in OrganicPharmaceutical Chemistry presented at Uppsala University in 2002

ABSTRACT

Schaal, W. 2002. Computational Studies of HIV-1 Protease Inhibitors. Acta UniversitatisUpsaliensis. Comprehensive Summaries of Uppsala Dissertations from the Faculty of Pharmacy263. 88 pp. Uppsala. ISBN 91-554-5213-2.

Human Immunodeficiency Virus (HIV) is the causative agent of the pandemic disease AcquiredImmune Deficiency Syndrome (AIDS). HIV acts to disrupt the immune system which makesthe body susceptible to opportunistic infections. Untreated, AIDS is generally fatal. Twentyyears of research by countless scientists around the world has led to the discovery andexploitation of several targets in the replication cycle of HIV. Many lives have been saved,prolonged and improved as a result of this massive effort. One particularly successful target hasbeen the inhibition of HIV protease. In combination with the inhibition of HIV reversetranscriptase, protease inhibitors have helped to reduce viral loads and partially restore theimmune system. Unfortunately, viral mutations leading to drug resistance and harmful side-effects of the current medicines have identified the need for new drugs to combat HIV.

This study presents computational efforts to understand the interaction of inhibitors to HIVprotease. The first part of this study has used molecular modelling and Comparative MolecularField Analysis (CoMFA) to help explain the structure-active relationship of a novel series ofprotease inhibitors. The inhibitors are sulfamide derivatives structurally similar to the cyclicurea candidate drug mozenavir (DMP-450). The central ring of the sulfamides twists to adopt anonsymmetrical binding mode distinct from that of the cyclic ureas. The energetics of this twisthas been studied with ab initio calculations to develop improved empirical force fieldparameters for use in molecular modelling.

The second part of this study has focused on an analysis of the association and dissociationkinetics of a broad collection of HIV protease inhibitors. Quantitative models have been derivedusing CoMFA which relate the dissociation rate back to the chemical structures. Efforts havealso been made to improve the models by systematically varying the parameters used togenerate them.

Keywords: HIV Protease, 3D-QSAR, CoMFA, Molecular Modelling, Force FieldParameterization, Quantum Mechanics, DFT, Enzyme Kinetics.

Wesley Schaal, Organic Pharmaceutical Chemistry, Department of Medicinal Chemistry,Uppsala University, Box 574, SE-751 23 Uppsala, Sweden

© Wesley Schaal 2002

ISSN 0282-7484ISBN 91-554-5213-2

Printed in Sweden by Uppsala University, Tryck & Medier, Uppsala 2002

Page 3: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

To Kaisa, Sonia and Ellen

Page 4: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

ABBREVIATIONS

3D-QSAR three dimensional quantitative structure-activity relationshipAIDS acquired immunodeficiency syndromeB3LYP Becke's 3-parameter exchange and Lee-Yang-Parr correlation functionalCADD computer-aided drug designCoMFA comparative molecular field analysisDFT density field theoryFF (empirical) force fieldHIV human immunodeficiency virusIN integraseKi inhibitory constantkon association ratekoff dissociation ratelogkon log10(kon)NNRTI non-nucleoside reverse transcriptase inhibitorNRTI nucleoside reverse transcriptase inhibitorpKi log10(1/Ki)pkoff log10(1/koff)PLS partial least squares or projections to latent structuresPR proteasePRI protease inhibitorQM quantum mechanicsQSAR quantitative structure-activity relationshipRT reverse transcriptaseSAMPLS sample-distance partial least squaresSAR structure-activity relationshipTS transition state

Page 5: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

PAPERS DISCUSSED

This thesis is based on the following papers:

I. Hultén, J.; Andersson, H. O.; Schaal, W.; Danielsson, H. U.; Classon, B.;Kvarnström, I.; Karlén, A.; Unge, T.; Samuelsson, B.; Hallberg, A. Inhibitors of theC2-Symmetric HIV-1 Protease: Nonsymmetric Binding of a Symmetric CyclicSulfamide with Ketoxime Groups in the P2/P2' Side Chains. J. Med. Chem. 1999,42, 4054-4061.

II. Schaal, W.; Karlsson, A.; Ahlsén, G.; Lindberg, J.; Andersson, H. O.; Danielson,U. H.; Classon, B.; Unge, T.; Samuelsson, B.; Hultén, J.; Hallberg, A.; Karlén, A.Synthesis and Comparative Molecular Field Analysis (CoMFA) of Symmetric andNonsymmetric Cyclic Sulfamide HIV-1 Protease Inhibitors. J. Med. Chem. 2001,44, 155-169.

III. Hämäläinen, M. D.; Markgren, P.-O.; Schaal, W.; Karlén, A.; Classon, B.; Vrang,L.; Samuelsson, B.; Hallberg, A.; Danielson, U. H. Characterization of a Set ofHIV-1 Protease Inhibitors Using Binding Kinetics Data from a Biosensor-BasedScreen. J. Biomol. Screen. 2000, 5, 353-360.

IV. Schaal, W.; Markgren, P.-O.; Hämäläinen, M. D.; Danielson, U. H.; Hallberg, A;Karlén, A. Comparative Molecular Field Analysis (CoMFA) of the Association andDissociation Rate Constants of a Diverse Set of HIV-1 Protease Inhibitors. Inmanuscript.

Reprints were made with permission from the publishers

Page 6: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

CONTENTS

1 INTRODUCTION 7

1.1 Etiology of AIDS 7

1.2 Structure of HIV 9

1.3 Replication of HIV 10

1.4 Current Targets and Agents of Anti-HIV Chemotherapy 13

1.5 Results of Anti-HIV Chemotherapy 17

1.6 HIV Protease 21

2 COMPUTATIONAL CHEMISTRY 24

2.1 Quantum Mechanics 24

2.2 Molecular Mechanics 27

2.3 Quantitative Structure-Activity Relationship (QSAR) 28

3 AIMS OF THE PRESENT STUDY 31

4 CYCLIC SULFAMIDE-BASED HIV PROTEASE INHIBITORS 32

4.1 Cyclic Urea-Based Inhibitors 32

4.2 Cyclic Sulfamide-Based Inhibitors 34

4.3 Study of the Ring Flip 35

4.4 Generality of the Ring Flip 38

4.5 Exploitation of the Ring Flip 40

5 KINETIC ANALYSIS OF HIV PROTEASE INHIBITORS 45

5.1 The Technology of Surface Plasmon Resonance Biosensors 45

5.2 An SPR Screen of HIV Protease Inhibitors 47

5.3 Analysis of the Screening Data 49

5.4 Quantitative Structural Analysis of Kinetics Data 50

5.5 Combinatorial CoMFA 52

5.6 Computational Details 54

6 EMPIRICAL FORCE FIELD PARAMETERIZATION 56

7 CONCLUDING REMARKS 62

8 ACKNOWLEDGEMENTS 63

9 REFERENCES 65

Page 7: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

7

1 ACQUIRED IMMUNODEFICIENCY SYNDROME (AIDS)

In mid-1981, five cases of a rare form of pneumonia (Pneumocystis carinii) and severe

viral infections in previously healthy young adults was rather quietly reported in Los

Angeles.1 Soon, an additional 26 cases of the pneumonia and another unusual disease,

Kaposi's sarcoma,2 were discovered in California and New York.3 The disease was

accompanied by a depressed immune system and a susceptibility to opportunistic

infections. This disease is now known by the name of acquired immunodeficiency

syndrome (AIDS).4-6

In the early 1980's, it would have been difficult to anticipate the full scope of AIDS

which by December 2001 has claimed the lives of 24.8 million. It has become the

leading cause of death in sub-Saharan Africa and fourth worldwide. Today AIDS is

recognized as a global epidemic which is not limited to any specific subpopulations.

With an estimated 40 million people currently infected and significant increases

expected in some areas of the world, AIDS could be classified as one of the worst

diseases ever known.7

1.1 ETIOLOGY OF AIDS

The first AIDS patients all had had a history of cytomegalovirus infection so this

became the first hypothesis of the origin of the disease8 but this was eventually

rejected.9 Other theories surrounded the fact the that the patients also fit a particular

demographic: homosexual males. The sexual stimulants amyl- and isobutyl nitrate

were implicated as possible etiological agents.10 This too could be quickly disproved

after similar cases were discovered in Haiti11 and Africa12 and among hemophiliacs,13

infants14 and women.15 Outside the scientific literature, theories regarding the cause of

AIDS have varied wildly to even include divine punishment.16

Beginning with the isolation of a novel retrovirus in 198317 which was later associated

with AIDS,18 a clearer picture of the disease began to emerge.9 This virus which has

Page 8: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

8

been known as HTLV-III (Human T-cell Leukemia Virus Type 3) and ARV (AIDS

related virus) is now known as human immunodeficiency virus (HIV).5,6 After some

initial resistance, HIV is generally agreed to be the sole causative agent though some

dissent remains in the scientific community even today.19

Two distinct types of HIV have been identified: HIV-1 and HIV-2.20 HIV-1 has been

further divided into three virus groups: the predominant M group, which is responsible

for most of the epidemic, and N and O.21 The origin of HIV-1 is most likely a cross-

species transmission (zoonosis) of a Simian Immunodeficiency Virus (SIV) from a

subspecies of Chimpanzee (Pan troglodytes troglodytes)22 but probably from different

events for each group.23-25 The date of the zoonosis events have not been precisely

discovered but antibodies against group-M HIV-1 were found in a serum sample

collected in the Belgian Congo in 1959.26 Models of the genetic divergence of the 11

subtypes of group M date a common ancestor somewhere around 1915-1941.27 HIV-2

is thought to have originated from zoonosis events from sooty mangabeys (Cercocebus

atys).28

The primary target of HIV seems to be CD4+ T lymphocytes which are part of the

machinery of our immune system.29 The primary phase of HIV infection progresses

fairly rapidly and may exhibit mononucleosis-like symptoms within a few weeks.30

During this early phase, the extent of infection is high and virion (virus particle)

concentration may exceed a million copies per ml blood.31 The host's immune

response usually kicks in after a few weeks and the level of virus in the blood declines

to bring HIV infection into its second phase. This long, asymptomatic period

characterizes HIV as a lentivirus ("slow virus").32 Viral replication is still active and

cells are rapidly being infected and eliminated during this period.33,34 The turnover of

T cells gradually leads to a decline in their number.35 In the third and final phase of

infection, the number of CD4+ T cells drops more quickly and the viral load increases

to produce clinical immunodeficiency.

Page 9: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

9

1.2 STRUCTURE OF HIV

Figure 1.1. Schematic of the HIV virion.

The mature HIV virion is an essentially spherical particle with a radius of about 10 nm

(Figure 1.1). The virus is surrounded by a lipid bilayer derived from the host cell and

contains several cellular membrane proteins.36 The outer portion of this envelope is

spotted with surface glycoprotein gp120 (named for its approximate molecular weight)

adhered to transmembrane protein gp41. The inside of the envelope is lined with

matrix protein p17. Within this shell is the conical capsid core made up of capsid

protein p24. The core holds two copies of the single stranded RNA which make up the

viral genome. As with all other lentiviruses, HIV is a retrovirus. This means that HIV

stores its genetic information as RNA which needs to "reverse-transcribed" to DNA.

Accompanying the genome are multiple copies of nucleocapsid protein p7, auxiliary

proteins Nef, Vif and Vpr and the essential enzymes: protease, reverse transcriptase

and integrase.37 Other auxiliary proteins, e.g. Vpu, Tat and Rev, are not thought to be

carried in the virion but are synthesized in the host cell.37,38

Page 10: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

10

1.3 REPLICATION OF HIV

A schematic representation of the replication cycle of HIV appears in Figure 1.2. A

myriad of cellular machinery is used to augment HIV's special tools.39,40 With over

175 000 articles indexed for HIV and/or AIDS on Medline,41 it is certainly one of the

most thoroughly studied systems today. As such, many details of the biology of HIV

will be omitted for the sake of brevity.

Figure 1.2. Schematic representation of the replication cycle of HIV.

Virus entry. The entry of HIV into a host cell may be divided into 3 distinct steps:

attachment, coreceptor interaction and fusion. Attachment of HIV-1 to the host cell

surface is mediated through gp120 on the virion surface binding to a CD4 antigen on

the host cell.42 Endogenous CD4 is present on the surface of many lymphocytes, which

make up a critical part of the body's immune system. This gp120-CD4 complex

interacts with a coreceptor on the cell surface, typically chemokine CXCR4 or

Page 11: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

11

CCR5.43 Transmembrane glycoprotein gp41 mediates membrane fusion to complete

virus entry into the host cell.

Uncoating the capsid core. Following fusion, the p24 encased capsid core is disrupted

to dump the contents into the cytoplasm of the host cell. It seems that this is

accomplished with the help of a cytoplasmic peptidyl-prolyl cis-trans isomerase called

cyclophilin A (hCyp-18) which had been incorporated into the virion.44,45

Reverse transcription. Successful entry of the contents of the viral capsid core is

followed by the reverse transcription of complementary DNA strand from the viral

RNA template by the viral enzyme reverse transcriptase (RT) in a complex with other

viral proteins.46 RT then degrades the RNA and produces the double-stranded viral

DNA. RT is highly error-prone since it is unable to catalyze the proof-reading which a

normal DNA polymerase performs.47

Nuclear import. The newly synthesized viral DNA is then imported into the nucleus of

the host cell. A short triple-helical region, made from a flap of about 99 bases,

synthesized during an interruption of reverse transcription seems to be necessary for

this event.48,49 Other viral proteins, such as Vpr,50 are also thought to be involved but

the system is complex.

Integration. The properly placed viral DNA is processed and transferred to the host

genome by the viral enzyme integrase (IN) as the central agent.46,51,52 Once the viral

DNA has been inserted, infection in that cell is for all intents and purposes permanent

since finding a way to selectively remove that little patch of DNA from the host

genome would seem to be a monumental task.

Transcription and translation. Once the viral DNA has been inserted into the host

cell's genome, HIV may persist in a latent, proviral state for many years in

unstimulated T cells.53,54 Activation of the host cells results in transcription of the viral

DNA by the host cell machinery into messenger RNA (mRNA). Early genes to be

Page 12: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

12

activated express auxiliary proteins Tat, Nef, Rev and a few others. Tat acts as a strong

promoter of viral transcription,55,56 Nef acts as a weak negative regulator57 and Rev

promotes switching to the expression of the structural proteins and enzymes.58

Regulation of viral expression involves a variety of interactions with the cellular

proteins.59,60 The auxiliary proteins have also been implicated in other roles such as the

down-regulation and degradation of cell-surface CD4 in infected cells by Vpu and

Nef, respectively, to promote the release of new virions.61-63

The second phase of transcription produces the unspliced mRNA for the precursor

proteins Gag (Pr55gag) and Gag-Pol (Pr180gag), which is the result of a translational

frame shifting event, in an approximately 20:1 ratio.64 The unspliced RNA is also

intended to be used as the genome of the next generation of the virus. Gag and Gag-

Pol are transported out of the cell nucleus by a poorly understood mechanism65 and

anchor to the wall through linkage with myristate at their N-termini.66,67

The precursor for the envelope glycoproteins gp120 and gp41 are treated like cellular

membrane proteins: synthesized, processed (glycosylated and cleaved) and transported

to the cell surface in the endoplasmic reticulum and the golgi apparatus68-70 though

some interaction with the Gag precursor has been implicated.71

Production of a new virion. Assembly of a new virus particle begins at the cell surface

with the clustering of roughly 2000 Gag proteins, 200 Gag-Pol proteins, processed

envelope proteins gp120 and gp41, two copies of the viral genomic RNA, some viral

tRNA and some other components like cyclophilin A which will be used after

infection of the next cell.72,73 It appears that the Gag protein mediates the budding

process.65 Some of the details involving cellular and viral components have recently

been elucidated.74-78 Release is assisted by viral protein Vpu in an incompletely

understood process.62

Virion maturation. The immature virion is a not-quite spherical blob with an outer

membrane derived from the host cell but including the viral coat proteins gp120 and

Page 13: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

13

gp41. The inside has roughly radial alignment of the Gag protein surrounding the

RNA79,73 though older work points to a more ordered structure.80 HIV protease (PR) is

required at this stage to cleave the Gag and Gag-Pol polyproteins into their constituent

structural (p17, p24, p7, p6, p2, p1) and functional (PR, RT, IN) proteins.81,82

External factors. The roles of outside factors has not been outlined here but they

certainly should not be discounted. For example, some narcotics have been shown to

act at least as cofactors in AIDS.83-85 On the other hand, coinfection with hepatitis G

virus (GBV-C) may actually improve chances for survival of AIDS.86-88

1.4 CURRENT TARGETS AND AGENTS OF ANTI-HIV CHEMOTHERAPY

This section will outline some of the chemical agents which are being used or

developed to combat HIV. Missing from this list are the most important tools of

education and modification of high risk behavior but these social issues are beyond the

scope of this thesis.89,7

First contact. The most attractive stage to halt is HIV before it enters the body.

Topical microbicides are being sought to prevent transmission. While some previously

promising candidates, e.g. the spermicide nonoxynol-9, have suffered doubts regarding

toxicity and effectiveness,90 other candidates are being tested.91 A similarly attractive

route is to develop a vaccine against HIV. While this wouldn't help those already

infected, an effective vaccine could be a safe way to halt the global spread of AIDS.

One potential vaccine is currently in phase III trials.92

Virus entry. A CD4 mimic (CD4-IgG2, PRO 542) which binds to gp120 to block HIV

attachment is in clinical trials.93,94 AMD-3100, which is currently in clinical trials, is a

CXCR4 antagonist which blocks the interaction with the gp120-CD4 complex.95-98

Other compounds are also under development for this target.99-101 In light of the recent

discovery of HIV strains which apparently can reproduce in CD8+ T cells in the

Page 14: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

14

absence of the CD4 antigen and CXCR4 coreceptor, it is possible that blocking this

target may not be completely effective.102

Peptides derived from gp41 have been found to interfere with its ability to initiate cell

fusion.103 Pentafuside (T-20, DP-178)104 and T-1249 (DP-107)105 are in clinical trials

and even a small, engineered protein is being investigated as an alternative.106

Uncoating the capsid core. Though compounds have been found to interfere with

cyclophilin A assisted uncoating,107 there is some doubt that useful drugs can be

developed since viral mutants which don't need to incorporate hCyp-18 have been

observed.108

Table 1.1. Nucleoside/-tide analog reverse transcriptase inhibitors

Name Code Approval a

zidovudine AZT/ZDV 1987

didanosine ddI 1991

zalcitabine ddC 1992

stavudine d4T 1994

lamivudine 3TC 1995

abacavir 1592U89 1998

tenofovir b PMPA 2001

emtricitabine FTC phase II/III

-- DAPD phase I/IIa Year approved for clinical use or current status in approval process. b Prodrug of a nucleotide

analog; all other compounds are nucleoside analogs.

Reverse transcription. Since this step is both essential and not duplicated by

endogenous enzymes, it has been one of the most active drug discovery targets.109 RT

inhibitors such as zidovudine (AZT) were the first clinically approved drugs for the

treatment of AIDS. Six nucleoside RT inhibitors (NRTIs) are currently clinically

available (Table 1.1): zidovudine,110 didanosine,111 zalcitabine,112 stavudine,113

Page 15: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

15

lamivudine114 and abacavir.115 At least two other compounds are currently in clinical

trials: emtricitabine116-118 and DAPD.119 These inhibitors function by being integrated

during reverse transcription and terminate viral DNA synthesis.120

A nucleotide analog, tenofovir (actually administered as disoproxil fumarate

prodrug),121 has been approved recently (Table 1.1). A nucleotide is a nucleoside

monophosphate. Since nucleosides must normally be converted to triphosphate form

(though some activity has been found for other metabolites of some analogs)122, a

nucleotide analog is expected to have the same sort of action as the nucleoside analogs

but have more rapid conversion to the active agent.123

Three members of a second class of RT inhibitors, the non-nucleoside RT inhibitors

(NNRTIs), have been approved (Table 1.2): nevirapine,124 delavirdine,125 and

efavirenz.126 At least four other compounds are currently in clinical trials:

emivirine,127-129capravirine,130 calanolide A131 and DPC-083.132 The NNRTIs don't

compete with nucleotide binding but interact at an allosteric site to block catalysis.133-

135

Table 1.2. Non-nucleoside analog reverse transcriptase inhibitors

Name Code Approval a

nevirapine BI-RG-587 1996

delavirdine U-90152 1997

efavirenz DMP-266 1998

emivirine MKC-422 phase III

capravirine AG-1549 phase II; on hold

calanolide A -- phase I

-- DPC-083 phase II/IIIa Year approved for clinical use or current status in approval process.

Page 16: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

16

Integration. Early inhibitors of IN with good in vitro activity failed to elicit sufficient

in vivo effect136 but new inhibitors are being developed137-139 and at least one, S-1360,

has entered phase II trials.

Production of a new virion. An inhibitor, MPI-49839, which may interfere with the

interaction between the p6 tail of Gag and endogenous Tsg101, which is normally

involved in membrane sorting, is being investigated in preclinical trials.140,141

Virion maturation. Inhibitors of PR have enjoyed a notable degree of success.142,143

To date, a total of six PR inhibitors (PI) are clinically available (see Table 1.3 and

Figure 1.7): indinavir,144 ritonavir,145 saquinavir,146 nelfinavir,147 amprenavir,148 and

lopinavir.149 At least four others are currently in clinical trials: atazanavir,150,151

tipranavir,152,153 mozenavir (Figure 4.2),154 and GW-433908.155,156 The current

collection of PR inhibitors bind in the active site but an alternative target is the dimer

interface (see section 1.6).157,158

Table 1.3. Non-nucleoside analog reverse transcriptase inhibitors

Name Code Approval a

saquinavir Ro 31-8959 1995

ritonavir ABT-538 1996

indinavir MK-639 1996

nelfinavir AG-1343 1997

amprenavir VX-478 1999

lopinavir ABT-378 2000

tipranavir PNU-140690 phase I/II

atazanavir BMS-232632 phase III

mozenavir DMP-450 phase I/II

GW-433908 b VX-175 phase IIIa Year approved for clinical use or current status in approval process. b GW-433908 is not a generic

name but just an alternate codename for VX-175.

Page 17: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

17

O NH

HN N

NH

HN O

O

O

OH O

O

N

Atazanavir

NH

SO O

N CF3OH

O

Tipranavir

Figure 1.3. Two HIV protease inhibitors in clinical trials.

Alternative agents. A number of natural products have been investigated formally and

informally.159-161 Often the mode of action is completely unknown and their

effectiveness is certainly questionable but at least one compound, the non-nucleoside

RT inhibitor Calanolide A, had reached clinical trials.131,162 The dangerous side of

unprescribed drugs has been documented in the interactions between an herbal remedy

for depression (St. John's wort)163 and prescribed HIV drugs.164 Problems associated

with drug-drug interactions have also been reported.165

1.5 RESULTS OF ANTI-HIV CHEMOTHERAPY

Zidovudine (AZT) was the recommended initial HIV therapy from its approval in

1987 to the mid-1990's. Zidovudine treatment significantly increased the chances for

survival for many patients but the effect was limited to no more than two years.166

After a few other NRTIs were approved in the early 1990's, combination therapy of

two drugs was found to have a greater effect on the progression of the disease. The

drugs zidovudine and lamivudine are synergistic since a common mutation which

confers resistance to one does not stop the other drug. While the side-effects of

zidovudine can be serious, e.g. anemia through bone-marrow suppression,167 they at

least weren't made worse in combination therapy.168

Page 18: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

18

Starting with the approval of saquinavir in 1995, PIs have been found to be effective

against NRTI-resistant HIV. The PI could act as salvage drugs but their greatest

impact was in triple combination therapy: typically two RT and one PR inhibitor.

Immune response improved and viral loads decreased dramatically even in patients

previously exposed to zidovudine. In the first three years after the introduction of this

therapy, the mortality rate dropped from about 30% per year to about 9% in the United

States. The effects were so successful that triple combination therapy is now referred

to as highly active antiretroviral therapy (HAART).169-172

Considering the clear benefits of HAART, it should be noted that HAART is used for

only a small minority of patients; the vast majority of infected individuals receive little

or no treatment at all. One factor is high cost, upwards of €10 000/person/year. Deals

have been made recently to lower the cost of the drugs by 90-99% to lower-income

nations but even this may be too expensive for the poorest. In addition, other issues

involving logistics, education and complex social factors must be resolved before

HAART can be a treatment for none but the patients in the wealthiest nations.173,174

Side-effects. Considering that AIDS was defined as a disease only about twenty years

ago, it shouldn't seem too surprising that the currently available drugs have at least a

few imperfections.175 The innately high toxicity of DNA chain terminators (NRTIs)

was certainly less significant back in the late 1980's when the alterative was rapid

disease progression which almost certainly lead to death. Now that HAART has kept

patients alive for years beyond initial estimates, the side-effects of antiretroviral

therapy have become both more significant and more apparent.

Probably the most serious side-effect is mitochondrial toxicity associated with NRTIs.

Mitochondria are the subcellular organelles which are responsible for generating the

energy cells need to function. Disruption of this function in any tissue leads to

catastrophic results when the energy demand exceeds the supply. The effect on the

liver or pancreas can be fatal. The mechanism is likely to involve inhibition of DNA

Page 19: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

19

polymerase γ to disrupt mitochondrial replication but other enzymes may also be

disrupted.176,177

Another important side-effect is Fat Redistribution Syndrome or Lipodystrophy.

Manifestations of the syndrome include loss/redistribution of body fat, leading to

significant changes in appearance, and disturbances to lipid/glucose metabolism,

possibly leading to insulin resistance and diabetes.178 It's commonly associated with PI

treatment179,180 but NRTIs may play a role (possibly through mitochondrial toxicity)

and the condition may arise in treatment-naïve AIDS patients.181 The mechanism of PI

associated lipodystrophy is under study.182

Cure? After a few months of HAART, plasma virus levels can drop to almost

undetectable levels. Analysis of viral decay rates initially suggested that eradication of

the infection might be possible within a few years.183 This estimate hinges on the

theory that an insignificant rate of viral replication would neutralize the infected cells

and the natural turnover of T cells would eventually purge the body of the infection.

Unfortunately, this goal has not been met184,185 as will be described below.

More careful detection techniques have shown that viral reproduction is not

completely suppressed under HAART.186-188 Continued viral replication leads to the

selection of viable mutant strains to eventually render the current drugs useless.

Reservoirs of latently infected cells are significant to the dynamics of HIV infection. A

significant pool of this type is the resting memory CD4+ T cells which form an integral

part of long-term immune response by "memorizing" antigens presented in the past.

The memory T cells generally remain in a resting state until an appropriate antigen

returns but activation can reinitiate infection at least in vitro and presumably in vivo.

These cells necessarily have a long half-life (about 44 months under HAART) and it

has been estimated that complete turnover could take over 60 years.189

Page 20: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

20

Reservoirs of unincorporated virus have been found in various tissues of the body. In

this state, the virus is immune to current therapeutic strategies since sometimes the

associated cells are not even infected but just have virions adhered to the cell surface.

Virus particles in this state have been found to still be capable of reinitiating

infection.190-192

Assuming that the enormous financial burden could somehow be overcome, the

prospect of 60 years of HAART may actually seem tolerable if one compares this to

traditional treatments for a disease like diabetes where injections and monitoring have

become a way of life. Since the side-effects of the current generation of HAART drugs

are quite serious and may lead to life-threatening conditions, this is not likely to be a

viable option. Furthermore, even the less deadly side-effects pose a very serious threat

in that it can help dissuade patients from faithfully maintaining the treatment; it can be

difficult to endure unpleasant side-effects when asymptomatic. Lapses in therapy can

lead to resurgence of disease and resistant strains may be less treatable both for the

patient and anyone later infected by this individual.

Future directions. Identification of the specific mutations selected by the current

drugs can help identify appropriate drug combinations and define the specificities

desired in the next generation of drugs.193,194 One way to get around the resting T cell

reservoir is to try to drain them during HAART.195 Early work on this seems

promising but not entirely effective.192 It seems reasonable that the reservoirs of

unincorporated virus would need to be taken care of too to really show a lasting effect.

It can be hoped that a new drug cocktail196 or a vaccine will come along soon to

completely cure the disease. For now, the only realistic alternative is continued

research into the current and new classes of drugs. Before that "magic cure" is

discovered, it is quite possible that future therapies will include an assortment of drugs

against HIV, drugs to bolster our natural response and drugs to counteract the side-

effects of the other drugs.

Page 21: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

21

1.6 HIV PROTEASE

The HIV protease (PR) was postulated to belong to the family of aspartic acid

proteases based on the identification of the Asp-(Ser/Thr)-Gly catalytic triad.202 Other

members of this family, including the endogenous enzymes Pepsin, Cathepsin D and

Renin, are single chain proteins of over 300 residues folded into two domains; each of

which supplies a catalytic triad of Asp-(Ser/Thr)-Gly. PR is much smaller at only 99

residues in length and possesses only a single Asp-Thr-Gly triad so a homodimeric

structure was proposed.203 Both of these conjectures were later confirmed by X-ray

crystallographic analysis of the apoenzyme204,200 and of a PR-inhibitor complex.205,206

a) b)

Figure 1.4. Ribbon drawings of (a) apo- and (b) inhibited HIV proteaseshowing the relatively open and closed position of the flaps (top of the images).These images were produced with MolScript197 and Raster3D198 on the PDB199

files 3HVP200 and 1AJX.201

The X-ray analyses revealed that the PR has C2 symmetry around a central active site.

The dimers are held together predominantly through interdigitated beta-sheets formed

at the base of the enzyme by the N- and C-termini of each monomer. The catalytic

cavity is covered by highly flexible flaps. In the apoenzyme (Figure 1.4a), these flaps

are in open position but when an inhibitor (or substrate) is bound (Figure 1.4b), the

flaps close down. The exact C2 symmetry of the enzyme can be broken upon the

binding of an inhibitor but the overall shape remains relatively consistant regardless of

the nature of the ligand (with the exception of very large molecules like fullerene

derivatives207).208

Page 22: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

22

The active site is a channel which has subsites for eight consecutive residues which in

the usual nomenclature210 are designated S4 to S1 before and S1' to S4' after the

scissile bond. The R-groups of the amino acids, or equivalent structures in non-

peptidic inhibitors, are designated P4-P4' to correspond to the appropriate subsites

(Figure 1.5).

N NH

HN

NH

HN NN

N

O

O

OH

OH

O

O

P1

P2

P3

P1'

P2'

P3'

Figure 1.5. A C2-symmetric HIV protease inhibitor (A-76928).209 P3-P1 andP1'-P3' represent the side chains intended to interact with the S3-S1 and S1'-S3'subsites, respectively.

The PR cleaves a variety of peptide bonds in the viral polyproteins during the course

of its action to produce the individual proteins of the mature virus. The active site

constellation of two proximal carboxyl groups from the Asp25/Asp25' residues (one

from each monomer) and a water molecule coordinated between the two carboxyl

groups are essential for catalytic activity. A hypothesis of the mechanism for peptide

bond cleavage by the PR is shown in Figure 1.6211,212 Recent studies213,214 have

challenged some of the details but the classical mechanism has been the starting point

for the design of many inhibitors incorporating transition state (TS) mimics based on

the putative tetrahedral intermediate shown in Figure 1.6 formed by the hydration of

the amide carbonyl group.215 All six of the currently approved PR inhibitors are

hydroxyethylene TS analogs, i.e., where the scissile amide bond is replaced by –

CH(OH)CH2–.

Page 23: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

23

NH

HN

O

R1

OHH

Asp25

O

O H

O

O

R2 NH

HN

R1

R2O OHH

O

O

O

OH

NH

HN

R1

R2O OHH

O

O

O

OH

NH

H2N

O

R1

R2

O

O H

O

O

δ-

OH

δ-

δ-δ-

Asp25' Asp25 Asp25'

Asp25 Asp25' Asp25 Asp25'

Tetrahedral Intermediate

Figure 1.6. Schematic representation of the catalytic mechanism of asparticacid proteases.

N NN

HN

OHN

OH

O

OH

NH

O

NHO

S

OH

OHN

NS

NN

S NH

HN

NH

O

O

O OH

O

O NH

O

NOH

S

O O

O

NH2

Indinavir

Nelfinavir

Ritonavir Amprenavir

HN

NHO

O

OHN

HN

O

H2NO

N

Saquinavir

OHN

NH

N NH

OO

OH O

Lopinavir

Figure 1.7. Clinically approved HIV protease inhibitors.

Page 24: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

24

2 COMPUTATIONAL CHEMISTRY

Computational Chemistry: A discipline using mathematical methods for the

calculation of molecular properties or for the simulation of molecular

behavior....216

Calculations for computational chemistry may be performed with anything from

massively-parallel super computers, desktop workstations, standard PC's or just a

pencil. Like so many other sciences, the dramatic increases in readily available

computational power has made some calculations considered too daunting to seriously

consider even 30 years ago seem almost routine today. For example, a bench chemist

who hits the "clean-up" button in a chemical sketching program would probably not

consider himself to be performing "computational chemistry": it's just too easy.

Working from this general definition, the field of computational is quite broad and

varied. Techniques of computational chemistry used in this study have included:

quantum mechanics, molecular mechanics, quantitative structure-activity relationship

(QSAR) analysis and experimental design. While these are in effect just tools, an

understanding of their working is hoped to provide a context for the chemically and

biologically relevant aspects of the present study.

2.1 QUANTUM MECHANICS

The familiar principles of Newtonian mechanics work amazingly well for most

anything directly observable. But when one inspects a system in minute detail, e.g., at

the atomic level, the simple equations aren't quite enough. Quantum mechanics (QM)

is a theory which states that there are discrete (quantized) levels of energy for a

system. The energy levels are so close together that the smooth functions of

Newtonian mechanics can be seen as an approximation of QM at high energies.

Page 25: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

25

The basic equation QM is the deceptively simple HΨ = EΨ, where H is the

Hamiltonian operator, Ψ is the wave function, and E is the energy. Actually, this is just

the short-hand form for the time-independent, non-relativistic Schrödinger equation

but the mathematical details are not really necessary here. In theory, the Schrödinger

equation should be able to describe nearly everything in chemistry (one needs to add

the relativistic mechanics of the Dirac equation to cover all of chemistry).

Unfortunately, exact solutions to the Schrödinger equation have only been found for

fairly trivial systems but the application of some approximations, chemically

interesting systems can be treated. One of the complications of the Schrödinger

equation is that the motion of the electrons and nuclear particles are coupled. Given

that the mass of a nucleus is thousands of times greater than that of an electron, their

relative motion can be approximately regarded as independent. This is called the Born-

Oppenheimer approximation and its application allows the electronic and nuclear

components of the Schrödinger equation to be solved separately. The electronic

component takes the greatest attention in QM but the nuclear component describes the

nuclear motions of spectroscopy as well as geometry optimizations. In a way,

molecular mechanics (Section 2.2) can be considered to work on the nuclear

component of the Born-Oppenheimer approximation.217,218

There are many other approximations which may be alternatively applied but from a

practical point of view, e.g., when using standard QM software, the two most

important issues to consider are which "level of theory" and which "basis set" to use.

Level of theory. With the application of a few principles of physics, like the Pauli

exclusion principle, QM in the context of chemistry (quantum chemistry) can be

divided to ab initio and semiempirical calculations of molecular systems. These

methods and their subtypes are often discussed as differing levels of theory where the

Schrödinger equation would be considered the generally inaccessible pinnacle.

Page 26: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

26

Ab initio roughly translates in this context as "from first principles" to denote that the

calculations are performed without experimental parameters. The basic, modern

implementation of ab initio is Hartree-Fock (HF).219 It basically extends a Born-

Oppenheimer type approximation to separately consider each wave function (Hartree's

theory) but tries to account for average field of electron repulsion (Fock's integrals).

Other commonly used methods, MP2 (second order Møller-Plesset perturbation

theory), MP3, etc treat electron correlation more accurately.220 This generally

produces better results but does so at a fairly high computational cost.221

An alternative approach to the MPn methods is Density Functional Theory

(DFT).222,223 While not strictly an ab initio method, since it includes a few empirically

derived parameters, it can achieve quite accurate results with only a modest increase of

computation time.221,224

Even with the use of limited basis sets and a moderate level of theory, ab initio

calculations can be quite computationally demanding. While this is certainly much less

of a problem today than even a few years ago, semiempirical methods225 greatly

expand the class of problems which can be studied. Semiempirical calculations

achieve their speedup by using a series of parameters to approximate the results of ab

initio calculations. Semiempirical calculations are frequently used today to calculate

approximate atom charges or to quick determine reasonably accurate geometries and

energies for many systems. They are also frequently used by QM software to "jump

start" ab initio calculations by calculating reasonable wavefunctions.

Basis sets. The ab initio methods can in principle be used to solve hydrogen atom

orbitals and then apply these solutions with their approximations to treat realistic

molecular systems. In practice, the orbitals are replaced with a series of approximation

functions, typically gaussians, which are collectively referred to as basis sets. Using a

large number of functions better approximates the real orbitals but (as expected)

increase the computational cost. Several standard basis sets are common usage, e.g.

STO-3G, 3-21G and 6-31G, but many other sets are available. Other choices are

Page 27: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

27

whether to add polarization (e.g., 6-31G* to add d orbitals) and/or diffuse functions

(e.g., 6-31+G* to also allow the orbitals to expand).221

2.2 MOLECULAR MECHANICS

A great simplification in molecular calculations is to simply ignore the motion of the

electrons and go back essentially to Newtonian mechanics. This is molecular

mechanics (MM) where an empirical force field (FF) describes molecular structure in

terms of average bond lengths, angles, torsions, etc and energetics in terms of force

constants. The force constants are restraining potentials generally approximated with

simple harmonic functions but sometimes with higher order terms. The FF is

parameterized to approximately reproduce various experimental results from

spectroscopy, calorimetry and possibly QM.

The chief advantage of MM is the incredible reduction in computational requirements:

both in computation time, on the order of several orders of magnitude, and memory.

This allows MM to be applied to systems which are impractical for QM.

The main limitation with MM is its dependence on the parameterization for accuracy.

For example, to properly simulate bond stretching, a good FF should: (i) provide

reasonable forces and distances for every combination of atom pairs within its

intended area of application (e.g., C–C, O–H, P–O, etc for biochemistry); (ii) account

for bond order (e.g., C–C versus C=C); (iii) consider immediate chemical environment

(e.g., a nitrogen of an amine doesn't act the same as a nitrogen of an amide); and (iv)

even considering effects of neighboring atoms (e.g., a carbons attached to the amide or

amine nitrogens will act differently). Add to this the combinations for three atoms

(bond angles) and then for four atoms (torsions) and the complication becomes clear.

A compromise must be made between accuracy and applicability (the transferability of

the parameters for one system to another). Many different FFs which have been

parameterized in different ways are currently in use. The different FF implementations

have somewhat different areas of applicability.226

Page 28: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

28

Procedures combining MM with QM have appeared. The best features of both can be

combined by treating some critical portion of the chemical system with QM and the

remainder with MM. This can be especially useful to simulate a chemical reaction

since bond breaking can't generally be simulated in MM; hard to follow the electrons

when none are present in the model.227,228

2.3 QUANTITATIVE STRUCTURE-ACTIVITY RELATIONSHIP (QSAR)

A basic premise at the core of medicinal chemistry is that similar structures can be

expected to exhibit similar biological activity. Formalization of this hypothesis into

"structure-activity relationship" (SAR) studies can help a chemist to design new

compounds with improved activity by analyzing the effects of substituents about a

common core. While counter examples can be found, this model is a basic tool of the

science.

A simple extension to SAR is to move from the qualitative to the quantitative (QSAR)

with the derivation of a mathematical formula to relate some quantifiable properties to

activity. These properties, often referred to as chemical descriptors, can be

experimentally derived or calculated quantities. Typically, QSAR studies focus on the

effects of a few substituents of a narrow congeneric series but the technique can be

generalized to use whole molecule properties like logP or polarizability.

Measurements of molecular properties can include descriptors based on their relative

three-dimensional properties. This technique, termed 3D-QSAR, has the potential to

be more interpretable since the descriptors can be more closely related to what a real

receptor feels. The greatest advantage of 3D techniques is their ability to move away

from congeneric series. Basically any combination of skeletons can be combined as

long as the molecules share the same mode of action at the receptor.

CoMFA. One popular application of the 3D-QSAR method is CoMFA (comparative

molecular field analysis).229 CoMFA works by calculating interaction energies of a

Page 29: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

29

probe atom with each compound of the dataset. The correlation between these

interaction energies and the measured biological activities is used to derive an image

of which regions in space are beneficial or deleterious to the activity.

To be directly comparable, the ligands must be aligned and oriented in their putative

bioactive conformations. Determination of this alignment rule is often considered the

most challenging aspect of CoMFA. Mutual alignment might be aided by

identification of a common pharmacophore using the active analog approach230 or

some other information. In fortunate circumstances, a ligand-receptor crystal structure

is available for guidance.

When all compounds in the data set have been superimposed they are located in a grid

box and the interaction energies (typically limited to the steric and/or electronic

interactions) between a selected probe atom and each molecule individually are

calculated at every lattice point in the grid box (Figure 2.1). This could be thought of

as painting a crude image of a receptor based on the 3D properties of the ligands.

Figure 2.1. The interaction energies between the probe atom and all moleculesare measured at each grid point on a regular 3D-grid. Each point in spacebecomes a descriptor variable in a QSAR analysis.

The steric (Lennard-Jones) and/or electrostatic (Coulombic) field energies thus

calculated become descriptors in the CoMFA table. The QSAR is generated by a PLS

Page 30: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

30

(partial least squares) analysis of the data contained in the table. PLS is a technique to

extract the principle components from a potentially large number of columns (the

interaction energies in CoMFA) in such a way to maximize the correlation with some

other value(s), e.g., biological activity. The statistical quality of the model can be

determined through the value of the crossvalidated r2 (q2). In crossvalidation, the

predictive ability of the model is estimated by repeatedly running PLS while leaving

out one (or more) compound(s) at a time until each compound is excluded. In each

round, the activity of the compounds that were left out is predicted. The q2 is computed

as a summary of the crossvalidation rounds and accumulates for each component of

the PLS. The number of components to use can be guided by the internal statistics of

the method which report a standard error of prediction. A model with a q2 below about

0.3 is probably unacceptable since that value could be found by chance correlation (the

exact cutoff value is a function of the number of compounds).231 A final CoMFA

model is then derived (without crossvalidation) using the optimal number of

components determined above to give a correlation coefficient (r2) for the model.

Page 31: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

31

3 AIMS OF THE PRESENT STUDY

This investigation is part of a research project aimed at the development of novel and

specific HIV-1 protease inhibitors. The specific objectives of this study have been:

(i) To elucidate the binding mode preferences of a cyclic sulfamide-based series of

HIV-1 protease inhibitors.

(ii) To derive useful structure-activity models of the cyclic sulfamide-based

inhibitors to aid future synthetic efforts.

(iii) To derive similar models based on the individual components of affinity,

namely association and dissociation rates, for a more diverse set of HIV-1 protease

inhibitors.

During the course of these studies, a new objective was identified:

(iv) To derive an accurate set of empirical FF parameters for sulfamide derivatives

in order to facilitate more accurate molecular modelling.

Page 32: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

32

4 CYCLIC SULFAMIDE-BASED HIV PROTEASE INHIBITORS

This chapter includes a background and summary of computational details of

Papers I and II of the complete thesis.232,233

Figure 4.1. Structural water-301 hydrogen bonding to the backbone NH's ofIle50 and Ile50' and the carbonyl oxygens of a linear inhibitor (BEA-322).234

4.1 CYCLIC UREA-BASED INHIBITORS

X-ray analysis of complexes of HIV PR with linear PIs generally include a tightly

bound, structural water molecule (Wat-301) bridging the enzyme flaps to the inhibitor

through hydrogen bonds to the Ile50/Ile50' amide hydrogens and the P1/P1' carbonyl

oxygens of the inhibitor (Figure 4.1). Hoping to reap entropic gains, researchers from

DuPont-Merck Pharmaceuticals looked for a way to incorporate that conserved water

into an inhibitor. Lead compounds were identified in a 3D database search and

developed into a distinctly non-peptidic series of cyclic urea-based inhibitors including

DMP-323 (Figure 4.2a).235 DMP-323 entered clinical trials but was later withdrawn

due to undependable bioavailability associated with its poor water solubility. Another

Page 33: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

33

member of this series, DMP-450, also known as Mozenavir (Figure 4.2b), has

substantially improved solubility and is currently in clinical trials.154,236

NN

HO OH

O

DMP 323

NN

HO OH

O

NH2H2N

DMP 450

HO OH

a) b)

Figure 4.2. Cyclic urea-based HIV protease inhibitors (a) formerly or (b)currently in clinical trials.

Figure 4.3. HIV protease inhibitors AHA-001 (black) and A-76928209 (grey)taken from the X-ray coordinates of protease-inhibitor complexes 1AJX and1HVK, respectively. Alignment was performed by superimposition of thebackbone atoms from each complex.

Our laboratory had also started to explore Wat-301 mimics237 but following the

publication of DMP-323, interest was drawn towards the design and synthesis of

cyclic urea derivatives.238,239 The parent compound of this series, AHA-001 (Figure

4.4a), has been co-crystallized with PR and the coordinates are available from the PDB

as entry 1AJX.201 The synthesis of AHA-001 and its derivatives is based on mannitol

(though other diastereomers have also been synthesized). The sugar sets the four chiral

Page 34: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

34

centers and the P1/P1' phenoxymethylenes extend a bit beyond the benzyls of DMP-

323. The overall binding mode of AHA-001 mimics that of DMP-323 which in turn

mimics the binding mode of many peptide-based linear inhibitors. Figure 4.3 shows

the similarity in binding of AHA-001 to a C2 symmetric linear inhibitor, A-76928

(Figure 1.5).209 Cyclic urea AHA-001 overlaps the P2, P1, P1', P2' and vicinal diol

(transition-state mimic) of A-76928. Also shown is the proximity of the carbonyl

oxygen of AHA-001 to the conserved structural water (Wat-301) associated with A-

76928.

4.2 CYCLIC SULFAMIDE-BASED INHIBITORS

A number of other mimics for water-301 have been incorporated into inhibitors

designed and synthesized by other research groups including phosphordiamidate,240

sulfoxide,241 sulfone,242 sulfamide,243 guanidine,244 oxamide245 and azalactam.246

Much of the work of the present study has focused on the cyclic sulfamide-based

inhibitors.238 A brief note on nomenclature: sulfamide derivatives (R2NSO2NR2) are

related to ureas (R2NCONR2) just as sulfonamides (R2NSO2R) are related to amides

(R2NCOR).

NSN

HO OH

O ONN

HO OH

O

OO OO

S2 S2´

S1 S1´ S1

S1´

S2´

S2

AHA001 AHA006

(a) (b)

Figure 4.4. Cyclic urea- and sulfamide based HIV protease inhibitors from ourlaboratories. The enzyme's subsites are marked to indicate, according to X-rayanalysis, where the side-chains have been directed.

Page 35: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

35

The parent compound of this series, AHA-006 (Figure 4.4b), is chemically identical to

AHA-001 except for the replacement of the water mimic. We expected that this

compound would adopt a binding mode similar to that of AHA-001. Preliminary X-ray

results based on this assumption showed strong distortions in the P1'/P2' arms of

AHA-006. Since it looked like the apparent (by analogy to AHA-001) P2' was too long

and the P1' was too short, maybe those groups have somehow switched positions.

Molecular mechanics calculations on the preliminary X-ray coordinates using

MacroModel 4.5247 were set up to test this hypothesis. The P1', P2' and the their

attachments in the central ring were allowed to relax while remaining ring atoms of the

central ring, P1 and P2 were constrained by a strong potential (100 kcal/mol). A short

calculation in vacuo under the AMBER* FF248 supported the hypothesis by quickly

switching the positions of what had been assumed to be the P1' and P2' (Figure 4.4).

Reanalysis and refinement of the X-ray data down to a 2.0 Å resolution showed a

decidedly nonsymmetric twist in the central ring associated with a switch of the P1'/P2'

side chains relative to AHA-001.201 The superposition of AHA-001 and AHA-006

from their protease complexes is shown in Figure 4.5. The phenoxymethylene groups

of each are colored black to illustrate the differences in binding conformation. The

inhibitors show pretty good overlap on the P1/P2 side where both phenoxymethylene

fit into S1. On the prime side, Figure 4.5 shows that the phenoxymethylene of AHA-

006 is placed into the S2' pocket and reaches further in than the benzyl of AHA-001.

4.3 STUDY OF THE RING FLIP

The X-ray analysis of one complex, even at good resolution (2.0 Å), may not be

enough evidence to be certain that the same binding mode would be adopted by the

whole series of compounds. We broke down the problem into two related sub

problems: (i) Is the flip induced by enzyme binding or is it a favored conformation in

bulk solution? In other words, is the twisted ring conformation the cause or result of

switched P1'/P2' groups? (ii) Regardless of the cause of the flip, could it be controlled?

Could we force a symmetric binding mode similar to the one seen in the urea

Page 36: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

36

derivatives by appropriate substitutions in the sidechains or, conversely, can we

depend on the flip to be there when we model new compounds? The latter question

will be addressed in Section 4.4.

Figure 4.5. HIV protease inhibitors AHA-001 and AHA-006 taken from the X-ray coordinates of protease-inhibitor complexes 1AJX and 1AJV, respectively.Alignment was performed by superimposition of the backbone atoms from eachcomplex.

The Cambridge Structure Database (CSD)249 was searched for sulfamides to address

the first question. The closest match was a seven-membered cyclic sulfamide ring with

a fused benzene ring in place of the diol (Figure 6.1; CSD code: SIKFUN).250 The

fused benzene ring certainly has an effect on the geometry but the sulfamide portion of

the ring matched qualitatively well with AHA-006 to lend some small bit of evidence

in favor of the idea that AHA-006 could form its geometry in solution (Figure 4.6a).

We have also tried to address the first question with molecular modeling by

calculating the energy difference between the nonsymmetrical and symmetrical

conformations. For the sake of simplicity, the twisted and symmetrical conformations

of AHA-006 were modelled with the side chains truncated to methyls (Figure 4.6b).

The starting geometry for the twisted conformation was taken from X-ray. The

symmetrical conformation was modelled with MacroModel 5.5 using AHA-001 as a

template. Restraining potentials on the side chains and diol were used and gradually

Page 37: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

37

diminished to guide the geometry to a stable point. Both model compounds were

relaxed to their nearest local minima.

a) b)

Figure 4.6. (a) Superimposition of the sulfamide ring of SIKFUN250 (black)and AHA-006 (grey). In this drawing of SIKFUN, the fused benzene oppositethe sulfamide was omitted and ethyl acetate in position 3 was truncated tomethyl. (b) Superimposition of the modelled symmetric (black) and observednonsymmetric (grey) conformations of the central ring of AHA-006.

In MacroModel 5.5, two FFs, AMBER* and MMFF94,251 were considered good

candidates since they could be used for the inhibitor in vacuo and latter to model the

protein-inhibitor complex. AMBER* reported low quality parameters for bond stretch,

angle and torsion terms involving the sulfur (actually, S-N-C-C torsions were reported

to be high-quality parameters even though the force constants were all zero). Attempts

to augment the parameters for sulfamide will be presented in Chapter 6. MMFF faired

better by reporting at least medium quality stretch and angle terms but many torsions

involving the sulfamide moiety (and elsewhere) were of low quality. Minimization

(without conformational analysis) from the X-ray coordinates by MMFF produced no

gross changes to the geometry. Considering this, MMFF was used for the general

modelling but molecular mechanics was judged to be unsuitable for the comparative

energy calculations.

Page 38: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

38

Semiempirical methods were considered briefly for the energy calculations but doubts

regarding parameterization252 prompted us to turn to ab initio calculations. The choice

of an appropriate basis set is generally important since it can strongly effect the

accuracy of the results or, just as easily, consume undue resources (see Section 2.1 for

a review). Mó et al253 have suggested that 3-21G* is minimally required for

sulfonamide but they and others (calculating on sulfonamide)254 have generally used

the 6-31G* basis set with the conclusion that larger basis sets added little accuracy

(relative to their experimentally derived values). The other consideration is what level

of theory to use. These researchers used Hartree-Fock (HF) but correlation effects with

levels of Møller-Plesset perturbation theory (MP)220 have also been explored for

sulfonamide and sulfamide.254,252 The opinions were mixed on the necessity of MP

considering their great computational cost so a compromise was chosen: density

functional theory (DFT). DFT calculations run at about the speed of conventional HF

but account for some electron correlation effects like MP.221 The B3LYP hybrid

functional255,256 was chosen as the specific implementation of DFT based on it's

generally good reputation.221

Geometry optimization using B3LYP/6-31G* was performed using Gaussian94257 to

find that the nonsymmetric conformation was favored for the model compound by 10

kJ/mol (2.4 kcal/mol). These energetic calculations support the interpretation of the X-

ray data and imply that the flipped conformation is achievable outside the protease

active site (with the caveat that only two conformations were studied). The 10 kJ/mol

energy difference also gives a hint for the second problem: the energy difference may

be surmountable within the enzyme with proper substitution of AHA-006.

4.4 GENERALITY OF THE RING FLIP

Well satisfied that the observed flip was at least reasonable, the second question of this

section of the study remains: Could we design compounds which would adopt a

symmetric binding mode or is the flip dependable? Six derivatives of AHA-006

(Figure 4.7) were designed to have good to strong preferences for the S2/S2'

Page 39: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

39

subsites258 which would be satisfied only if they would adopt a symmetrical, "urea-

like" conformation. A modelling study of these compounds was instituted to address

this problem.

NHO OHSN

HO OHO O

O O

AHA021

NSN

HO OHO O

O O

O O

O O

AHA019

NOH

NHO

NSN

HO OHO O

O O

AHA030

NSN

HO OHO O

O OOO

AHA025

NSN

HO OHO O

O O

OH OH

AHA023

NSN

HO OHO O

O O

O

O

O

O

AHA022

Figure 4.7. Compounds synthesized to test sulfamide ring flip hypothesis.232

Ambiguity regarding the Z/E configuration of the ketoximes of AHA-030 hindered its

modelling. Since it was predicted to be the compound most likely to force symmetrical

binding, its accurate modelling was important. A search of the CSD for acetophenone

oximes revealed 12 entries for E and 1 entry for Z. Energy calculations were carried

out to test the hypothesis of a more stable E isomer. An in vacuo conformational

analysis centered on the ketoximes and associated phenyl rings of AHA-030 was

performed with the MMFF force field of MacroModel 5.5. The E isomer was

calculated to be 14 kJ/mol (3.4 kcal/mol) more stable than the Z isomer. Since oximes

are not well parameterized in the version of MMFF used, a quantum mechanics

calculation was performed. The E and Z isomers from the MMFF calculation were

optimized at B3LYP/6-31G*. The E isomer was found to be 11 kJ/mol (2.6 kcal/mol)

more stable. With the assumption that the synthetic reaction used wasn't

overwhelmingly kinetically controlled in the Z direction, we used the E isomer in our

models.

With a working model for the AHA-030 ketoxime in hand, we then proceeded with the

modelling of the AHA-006 derivatives in both the symmetric and conformations

Page 40: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

40

nonsymmetric. Starting structures for each were taken from the model compounds

discussed in Section 4.3. Since the benzyl derivatives attached to the ring at the

nitrogens (i.e., the P2/P2' groups in a symmetrical conformation like AHA-001) were

the only portions which differed from AHA-006, we made the simplification in the

modelling work that the remaining portions of the inhibitors remained fixed. While

this assumption was far from rigorous, it did avoid the problem of inadequate

parameters in the empirical FFs mentioned in Section 4.3. Minimization and

conformational analysis centered on the benzyls and their attachments was performed

with the AMBER* FF and a GB/SA solvent model259 within the active site of the

protease from the AHA-006 complex.

The modelling predicted that the symmetric conformations of all derivatives would

allow favorable contacts with either Asp29/Asp29', Asp30/Asp30' or reach into the

solvent. Nonsymmetrical conformations would necessarily loose the S2' pocket's polar

interactions but they may be able to favorably interact with Arg8.

The derivatives were synthesized and tested against HIV-1 PR. By comparison to the

Ki values of similar cyclic urea derivatives made elsewhere, we concluded that the

most reasonable SAR would support the hypothesis that all of these derivatives

adopted the nonsymmetrical conformation observed for AHA-006. The X-ray structure

of AHA-030 in PR was determined and the nonsymmetric binding mode was clearly

visible. With this last bit of confirmation, we've concluded that the nonsymmetric

binding mode seems to be reproducible and robust.

4.5 EXPLOITATION OF THE RING FLIP

Convinced of the dependability of the nonsymmetric conformation, we decided to

make a series of chemically (rather than merely conformationally) nonsymmetric

derivatives which we hoped would be better adapted to the binding sites. While the S1'

and S2' pockets share similar characteristics, they are certainly not equivalent.260 A

good P1' group cannot be expected to be optimized for the S2'. This is illustrated in

Page 41: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

41

Figure 4.5 with the parent compounds AHA-001 and AHA-006: note how much

further AHA-006 reaches into the S2'. These differences in SAR should be understood

and exploited to improve the binding of the cyclic sulfamide inhibitors.

NSN

HO OHO O

O ONH

O

AHA047

HO ONSN

HO OHO O

O O

O

AHA024

NSN

HO OHO O

O ONH

O

NH

O

AHA045

Figure 4.8. Two of the chemically nonsymmetric AHA-006 derivatives (AHA-024 and AHA-047) synthesized to take advantage of the expectednonsymmetric binding mode and one of the symmetrical derivatives (AHA-045)for comparison.233

Eleven chemically nonsymmetric derivatives were synthesized along with seven new,

symmetric derivatives to help with the interpretation of the SAR (a few representative

structures appear in Figure 4.8). With the advantages we hoped to gain with this

chemical asymmetry, we received the complication that we can't a priori determine

which side of the inhibitors will adopt the flip: left or right. Because of this,

conventional SAR was not readily interpretable. We decided to use a quantitative

method to model which flip had been adopted by each nonsymmetric compound. The

decidedly three-dimensional nature of the problem prompted us to consider 3D-QSAR.

Many other QSAR261 and 3D-QSAR studies262-269,233 have been made on HIV-1

protease but this study was intended primarily to model the choice of ring flip.

As noted in Section 2.4, the relative alignment of the compounds is of critical

importance in 3D-QSAR. Fortunately, the crystal structure of AHA-024 complexed

with HIV-1 PR was solved to 1.8 Å resolution and available (PDB code: 1G35) to aid

in the alignment of the nonsymmetric compounds. Incidentally, the X-ray structure

showed a twisted ring conformation and the flipped P1'/P2' side chains in agreement

with AHA-006 and AHA-030.

Page 42: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

42

One flip conformation was known but the problem of the others remained.

Conformation-independent techniques have appeared in the literature270,271 but

considering the availability of good crystal structures to guide the fit of our congeneric

series, we wanted to take full advantage of the information we had. Another variation

is 4D-QSAR which can consider many conformations simultaneously.272 Since we

only wanted to consider two conformations per nonsymmetric compound, the extra

complications involved in 4D-QSAR (i.e., genetic algorithms) seemed unnecessary. In

the end, we opted to use standard CoMFA229 to generate models for all possible flip

combinations of the nonsymmetric compounds. With eleven nonsymmetric

compounds (considering AHA-024 as a control) in two different binding modes we

need 211, or 2048, CoMFA models. Exploration of the most suitable set of CoMFA

parameters would not be easy to achieve for this many models so we settled for the

very limited survey of only the ten fields offered in the Advanced CoMFA package. In

total, we now had 20480 models to generate.

Running 20480 CoMFA calculations interactively would at the very least be terribly

boring so these calculations were run in batch mode using a script. The details of the

procedure, along with a more complex CoMFA analysis, will be presented in Section

5.6.

To prepare the dataset for CoMFA, minimization and conformational analysis using

AHA-006 and AHA-024 as templates was set up and performed basically as described

in Section 4.3, with the exception that only the nonsymmetric conformations were

being considered. The eleven nonsymmetric compounds were modelled in both flip

conformations. AHA-006 and the six derivatives from the previous study were also

included in the models to bring the total to 25 compounds in each model.

The q2 (crossvalidated correlation coefficients) values from the CoMFA calculations

were used as a rough measure of the quality of the alignment (binding mode) of the

nonsymmetric inhibitors. The numerically sorted q2 values from these models (Figure

4.9) form a normal distribution curve with a few values peaking above 0.7. The top 20

Page 43: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

43

models were considered carefully in the context of molecular modelling. The model

with second highest q2 was eventually chosen though several other models were just

about as reasonable. This ambiguity could be rationalized in several ways but the most

significant may be that some of these inhibitors may be able to bind almost equally

well in either flip conformation. The CoMFA calculations could conceivably be

modelling this accurately but this may be pushing the data a bit too far.

Figure 4.9. Crossvalidated correlation coefficients (q2) for 20480 CoMFAmodels (sorted by q2).

Before publication of these results, the crystal structure of AHA-047 complexed with

HIV-1 PR was solved to 1.95 Å resolution (PDB code: 1G2K). The X-ray structure

again showed a twisted ring conformation and the flipped P1'/P2' sidechains in

agreement with the other sulfamides. The twist was seen to lie on the side of the

unsubstituted benzyl in agreement with the conformation in the chosen CoMFA

model.

As stated, the purpose of the CoMFA calculations was really just try to figure out the

binding modes; to help make a reasonable guess as to which flip each nonsymmetric

inhibitor might adopt. But with a guess of the alignment, we started a more

conventional CoMFA calculation on the dataset. For this calculation, we used the 18

Page 44: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

44

compounds of the current study233 as a training set and the seven old compounds (the

six from the previous study232 plus AHA-006) as a test set. The resulting model (q2 =

0.54, r2 = 0.96, 3 components) predicted the test set reasonable well with a mean

absolute residual pKi of 0.58.

Page 45: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

45

5 KINETIC ANALYSIS OF HIV PROTEASE INHIBITORS

This chapter includes a background and summary of computational details of

Papers III and IV of the complete thesis.273,274

In the early phases of drug development, an understanding of the details of receptor

interaction and of the ADME (absorption, distribution, metabolism and excretion)

profile of the candidates can be of great importance.275,276 But regardless of how well

the system is understood, some information regarding the affinity for the target must

be ascertained. In this age of high-throughput screening,277 this information might

come in the form of a binary, yes/no answer instead of a precise value possible with

"low throughput" Ki determinations.

An intermediate method which can be used to complement these two extremes are the

aptly named moderate-throughput screening methods. The instrument which has been

used in this study is a surface plasmon resonance (SPR) based biosensor. One of the

advantages of using this sort of instrument is that the affinity measurement, KD, is

broken up into its constituents of association rate (kon) and dissociation rate (koff). The

significance of having access to this extra information will be discussed in Section 5.4.

5.1 THE TECHNOLOGY OF SURFACE PLASMON RESONANCE BIOSENSORS

When polarized light is shown through glass onto a thin metal film, there is a dip in the

intensity of the reflected beam at a specific angle of incidence. This angle is sensitive

to the refractive index at and near the surface.278 When molecules bind to that surface,

the refractive index changes and this, in turn, changes the angle for maximum

absorbance. Detecting this change of angle over time is at the heart of an SPR sensor.

When biologically interesting molecules are immobilized onto this surface, e.g. a

protein or antibody, it becomes a biosensor which can detect molecular binding in real-

time without the need for fluorescent or radioisotopic labels.279-281

Page 46: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

46

The generalized schematic of a flow cell in Figure 5.1 illustrates the essential

components of an SPR biosensor. Several of these flow cells may be within an

instrument to allow the simultaneous detection of the same substance binding to

several different immobilized targets. Subtraction of the signals across the detectors,

e.g. one for specific binding to a receptor and another for non-specific binding to

albumin, could reduce the errors associated with subtracting the final values from

separate experiments. Alternatively, different substances, e.g. reference and test

compounds, could be run in each channel against the same type of target to achieve

similar benefits.

Figure 5.1. Schematic diagram of a surface plasmon resonance biosensor.

The SPR signal is expressed as resonance units (RU) and the continuous display of RU

as a function of time is referred to as a sensorgram. The idealized sensorgram shown in

Figure 5.2a illustrates the basic stages of the instrument's cycle: (i) buffer blank pulse

used as a negative control and to detect carryover between samples; (ii) sample pulse

separated into association (during sample injection) and dissociation (after sample

injection) phases and used for identification of binders; (iii) regeneration of the sensor

surface used to remove slowly dissociating binders; and (iv) system wash to rinse the

autosampler and the injection unit. Injection of a sample that interacts with the sensor

surface results in a signal that, after subtraction of the reference signal, is proportional

to the amount of bound ligand. The dissociation phase starts at point D, once the

Page 47: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

47

injection has been switched from sample to running buffer. The rate of signal increase

during sample injection (starting at A) provides the observed association rate constant

(kon), while the observed dissociation rate constant (koff) is obtained from the rate of

signal decrease after point D.

a) b)

300

200

100

0

-100

Res

pons

e(R

U)

Time (s)

A

D

Bla

nkB

lank

Sam

ple

Sam

ple

Reg

ener

atio

nR

egen

erat

ion

Was

hW

ash

0 300 600

Figure 5.2. (a) Cartoon of response (RU) versus time for a cycle of an SPRbiosensor showing the stages of blank injection, sample injection, regenerationand wash. (b) Typical sensorgrams (truncated on the y-axis) for three HIV-1protease inhibitors. Reporter points for the association phase (A1 and A2) anddissociation phase (D1 and D2) are indicated.

5.2 AN SPR SCREEN OF HIV PROTEASE INHIBITORS

The development of biosensor technology has provided a new tool for rapid kinetic

studies of biomolecular interactions and recent improvements in sensitivity and

methodology allows the technique to be used for interaction studies with low

molecular compounds as analytes.281-283 The work in our laboratories has focused on

the interaction between HIV-1 protease and inhibitors.284-287

Page 48: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

48

The present study describes a screen of 290 HIV-1 protease inhibitors.273 These

structurally diverse compounds included both linear and cyclic inhibitors culled from

several cooperating laboratories. The structures covered a range of molecular weights

from 232 to 1093, MlogP (calculated Moriguchi logP) from -2.7 to 5.2 and possessed

from 3 to 20 hydrogen bond acceptors. Reliable inhibition data (Ki) was available for

all for comparison purposes. These values ranged from 70 pM to a cutoff of 10 µM for

inactive compounds.

Figure 5.3. Association phase reporter point A1 versus measured pKi (-log10Ki). The group of points with a pKi of 5 represent a cutoff of 10µM for Kiimposed on the data.

A typical sensorgram from three HIV PR inhibitors is shown in Figure 5.2b. The

reporter points for the association phase (A1 and A2) and dissociation phase (D1 and

D2) served as the primary data in the study where each compound was run at a single

concentration. Using one report point of the association phase (either A1 or A2), we

found a reasonably good correlation (r2 = 0.72) to pKi (Figure 5.3). It should be noted

that this correlation overcomes the fact that the buffer for SPR284 used only 0.15 M

NaCl which is quite different than the 1 M NaCl typically used for the Ki239

determinations.288

Page 49: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

49

5.3 ANALYSIS OF THE SCREENING DATA

The results presented in Section 5.2 relied on processing the raw data from the

sensorgrams with the instrumental software. At that point, there was still a large

amount of data to consider for a screen of 290 compounds. Besides the four reporter

points, there was data for the washing and regeneration stages as well as correlations

to the results from the other channels besides HIV PR.

The processing of this data was aided by scripts written in the Perl programming

language289 to filter out dubious values and find reasonable thresholds for some of the

variables to achieve the high correlation of biosensor data with previously measured

activities. An example of the data processing which occurred in the background is

given below.

a) b)

Figure 5.4. Association data for report point A1 expressed as % response of thereference inhibitor (Indinavir). (a) Data arranged alphabetically by substancecode name. (b) Same data (negative values removed for clarity) reordered bydate of experiment. Individual experiments are separated by the marks belowthe x-axis.

Figure 5.4a shows some early results for A1 expressed as the relative % response of

Indinavir on the y-axis. At this stage in the investigation, most compounds had been

measured in duplicate and all data points for each compound are shown in the figure.

Page 50: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

50

A completely unexpected results was the number of compounds which expressed very

high association data. Over 10% of the measurements reported A1 values over 200%

of the average Indinavir signal which would roughly correlate to a Ki in the high

femtomolar range. That estimate is from an extrapolation far beyond the range later

determined for the full dataset (Figure 5.3) but it's good enough to indicate a problem

since none of the compounds from this 10% had measured Ki values much below the

nanomolar range.

Reordering of the data by the time of the experiment (Figure 5.4b) showed a definite

pattern: the second and third experiments produced the suspicious results.

Identification of this troubled region of the dataset allowed us to identify the specific

problem (high refractive index of the bulk solution in two plates). Simply eliminating

all data with high A1 and A2 values would certainly not be reasonable since we would

have to presuppose that nothing significantly better than Indinavir could be found.

Assuming that all duplicates with a small relative error to be dependable would also

have been wrong since some of the bad data had been duplicated. Elimination of the

association data for these two experiments was certainly the most prudent course of

action.

5.4 QUANTITATIVE STRUCTURAL ANALYSIS OF KINETICS DATA

The commonly reported steady-state inhibition constant, Ki, is a standard (though not

exclusive) determinant for whether a compound can become a lead or rejected. The

related equilibrium dissociation constant (KD) is a composite term of the association

(kon) and dissociation (koff) rates: KD = koff / kon. These rates are independent quantities

which not only describe different aspects of binding but behave differently in different

environments.290,291,288 The pharmaceutical importance of breaking affinity into its

constituent parts has recently been discussed in the context of HIV PR.292,287

The work presented in Section 5.2 was a broad screen of PR inhibitors so only a few

reporter points for association and dissociation were used. A more detailed study of a

Page 51: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

51

diverse subset of these compounds was selected for a more detailed kinetics study.293

In that study, the kon and koff values were used to make SAR analyses of the inhibitors.

Some other studies have noted correlations of kon and/or koff to some chemical

descriptor and the idea of using this in a QSAR study has been presented294 and

carried out.295,296 It is believed that the work presented in this chapter is the first

application of a 3D-QSAR technique, in this case, CoMFA,229 to the study of kon and

koff.

The dataset for the CoMFA study consisted of 34 compounds. These were split into

training (22) and test (12) sets via an experimental design based on several chemical

descriptors. It was hoped that the chemical design would help insure a good coverage

of some features of the chemical space without making biased selections.

The derived CoMFA model produced a reasonably good q2 of 0.44 for the dissociation

rate (Figure 5.5a). The model reproduced the test set data with a predictive correlation

coefficient (r2pred) of 0.59. The q2 for the association rate was a much less impressive

0.25 which was close to the point of being useless (Figure 5.5b).231 The test set gave

correspondingly poor r2pred of 0.14. A bit disappointed with these results, we embarked

on a campaign to find better models.

Figure 5.5. Plots of actual versus calculated (a) pkoff or (a) logkon for thetraining (crosses) and test (circles) sets of the CoMFA models with defaultsettings. The dashed-lines mark one-to-one ratio for reference only.

Page 52: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

52

5.5 COMBINATORIAL COMFA

CoMFA, as implemented in the Sybyl molecular modeling package,297 defines many

adjustable parameters. Many CoMFA studies have reported adjusting some of these

parameters to produce models of significantly better (and worse) statistical

quality.298,299 In the hopes of finding better CoMFA models for the association and

dissociation rates, we used the experience gained from the binding mode search

presented in Section 4.5: we used a script to automate the calculations. Instead of

varying the binding modes, we varied the CoMFA parameters.

Our first step was a short search to position the grid over the compounds. The default

grid was adjusted in 0.5 Å increments along the x, y and z axes independently. A total

of 189 grids were used. Leave-one-out validation was used for all models. The

improvement of q2 scores for koff gave a respectable 0.59 (up from 0.44) and the q2 of

0.37 for kon became at least acceptable. Still not satisfied with these results, the search

proceeded with other adjustable parameters. The five best grids (as ranked by their q2

score) were carried over to the next stage.

Table 5.1. Adjusted CoMFA parameters.

Variable Values

FIELD_TYPE ELECTROSTATIC, STERIC, BOTH

STERIC_ENERGY_MAX 80, 60, 45, 30, 15, 5

ELEC_ENERGY_MAX 80, 60, 45, 30, 15, 5

VOLUME_AVG_TYPE NONE, BOX

SWITCH_FCN NO, YES

HBOND_FCN NO, YES

TRANSFORM NONE, INDICATOR, SQUARED

The COMFA parameters adjusted in this search appear in Table 5.1. The variable

names correspond to those used in the "tailor comfa" settings of Sybyl and the

technical explanations for them can be found in the Sybyl manual.297 All of these

Page 53: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

53

variables correspond to settings which are accessible through the normal, interactive,

graphical mode of Sybyl (assuming the appropriate modules are available).

Figure 5.6. Plots of actual versus calculated (a) pkoff or (a) logkon for thetraining (crosses) and test (circles) sets of the CoMFA models with optimizedparameters. The dashed-lines mark one-to-one ratio for reference only.

Systematic variation of these variables, avoiding the disallowed or unproductive

combinations, resulted in the generation of 348 CoMFA models for each of the five

grid files and both koff and kon to give a total of 3480 CoMFA models, each with leave-

one-out validation. The q2 score for koff was now a very nice 0.72 (Figure 5.6a) and kon

improved to 0.48 (Figure 5.6b); higher than the default model for koff.

While q2 is a standard measure of the quality of a model, it certainly doesn't match an

external prediction of a test set. The r2pred for the test set against the improved model for

koff was 0.60. Compared to the default model's 0.59, this is certainly no real

improvement. The test set was reasonably well predicted for the default CoMFA

model so at least the model didn't do any worse after parameter optimization.

Substantial improvements were needed for kon which had a r2pred of 0.14 for the default

model. Unfortunately, the substantially improved model didn't fare much better: r2pred =

0.20. At least for this dataset, the significant improvements in q2 did not translate into

a tangible benefit for prediction of the test set.

These results (as some other studies have shown)263,300,299 should at least be a warning

against the over reliance on q2. For example, the q2 values for koff span a range from -

Page 54: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

54

0.27 to 0.72 for the various combinations of adjustable parameters (Figure 5.7). This

can be interpreted in at least two ways: (a) the default CoMFA settings are not

necessarily the best parameters for all models, or (b) a given set of parameters can

produce any of an incredibly wide range of values.

Figure 5.7. Crossvalidated correlation coefficients (q2) for the 1740 CoMFAmodels calculated for the dissociation data (modelled as pkoff).

It should also be stressed that the work presented here represents only a single dataset

(albeit with two y variables) so the generality for r2pred is far from certain. Further

research into this question for other datasets as well as a deeper analysis of the q2 data

is in progress.

5.6 COMPUTATIONAL DETAILS

Varying the CoMFA parameters combinatorially is a slow, "brute force" search.

Luckily, the calculation of 3480 models was accomplished in about 7½ hours. This

speed is partly thanks to the fast alternative to a full PLS301 calculation called

SAMPLS (Sample-distance Partial Least Squares)302. Sybyl Programming Language

(SPL) and UNIX shell scripts were written to manage the SAMPLS calculations.

Minor adjustments to the SAMPLS control script

Page 55: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

55

($TA_ROOT/comfadv/tables/sampls.core) were necessary since the global Sybyl

variables QSAR_STDERR and QSAR_CROSS_R2, reporting the standard error and

crossvalidated correlation coefficient (q2) for each component, were not being used.

The sampls.core script was altered to set these global variables to the local values of

std_err and cross_r2, respectively. The implementation of SAMPLS in Sybyl also

suffers from a small memory leak which becomes problematic after the calculation of

a few hundred CoMFA models. A UNIX shell script was used to restart Sybyl after

about every 100 CoMFA calculations.

Page 56: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

56

6 EMPIRICAL FORCE FIELD PARAMETERIZATION

This chapter is a brief description of some unpublished results of direct

relevance to Chapter 4 of this thesis. The preparation of a manuscript is in

progress.

As was mentioned in Section 4.3, the sulfamide moiety R2NSO2NR2 is not well

parameterized in the available empirical force fields. As a result of this missing data,

the molecular modelling studies of the cyclic sulfamide HIV-1 PR inhibitors (Figures

4.4b, 4.7 and 4.8) presented in Chapter 4 were quite limited. Basically, we relied on

some X-ray structures to align the inhibitors and allowed the sulfamide ring to explore

at most one alternative conformation (Figure 4.6b). While the available X-ray data has

so far indicated a single, consistant ring conformation, it is difficult to assume that no

other low energy conformations exist in solution.

The ability to accurately perform an extensive conformational analysis, as has been

reported for cyclic ureas,303 could help answer this question. Furthermore, unrestrained

energy minimization would open up the possibility of using some alternatives to

CoMFA for activity prediction.304 In the hopes of being able to eliminate these

limitations, work has been initiated to make new parameters for the AMBER* force

field248 of MacroModel.247

Several routines to fit new parameters for existing FFs have been described using

neural networks,305 simplex optimization306 or genetic algorithms.307 We have used the

procedure of Norrby and Liljefors.308 Their technique uses QM frequency calculations

at non-stationary points similar to the procedure309,251 used to derive some of the most

generally successful FFs in common use.226

In contrast to some of the more simple FF parameterization procedures,310 this method

allows the inclusion of experimental data. Crystallographic data for several sulfamide

derivatives (Figure 6.1) were used as models for energy conformations (listed here by

Page 57: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

57

CSD code): CITSON,311 CITSON10,312 DMAMSO,313 FIKHEM,314 FIKHIQ,314

GABGIZ,312 GABGUL,312 KIBRAO,315 KIKSEC316 and SIKFUN250. Spectroscopic

data for tetraalkylsulfamides was used as secondary data source.317-319

NSN

O

O O

CITSON &CITSON10

NS

N

O O

DMAMSO

NSN

OO

O O

Bu

FIKHEM

NSN

O

O O

GABGIZ

NS

N

O

O O

Br

GABGUL

NS

N

O O

N

Cl HN

NH

KIBRAO

SN

N

N

N

O

O

N

N

SN

N

O

O

KIKSEC

NSNO O

O

O

HOSIKFUN

Figure 6.1. X-ray structures used to guide force field parameterization.

DFT calculations. As done in Section 4.3, B3LYP/6-31G* with Gaussian94 was used

for the QM calculations. Tetramethylsulfamide (DMAMSO in Figure 6.1) was used as

the model compound for the cyclic sulfamide inhibitors like AHA-006 (Figure 4.4b).

Using a smaller model compound, e.g., unsubstituted sulfamide, might not correctly

reproduce the electronic character of the inhibitors. Using a larger molecule as the

model, e.g., the truncated sulfamide ring modelled in Section 4.3 (Figure 4.6b), would

be computationally more expensive and possible not sufficiently flexible to allow a

good fitting of the torsional data.

The individual iterations of the geometry optimization are shown in Figure 6.2a. The

small jumps in the energy near the beginning of the optimization are not strange and

even the large spike to 116 kJ/mol at the 34th iteration isn't so unusual. What is

troubling is that the optimization never converged. As seen in the expansion of the last

Page 58: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

58

part of the optimization run (Figure 6.2b), the energies are oscillating. In Gaussian94,

convergence for geometry optimizations are reached only the forces on the atoms

(maximum and RMS) as well their displacements (maximum and RMS) are below

some predefined limits. In this calculation, the forces were well below the cutoffs but

the structure couldn't stabilize. Since the oscillations were of such low amplitude, less

than 10 J/mol (in the microhartree range), this nearly optimized structure was used in

the next stage of the calculations.

a) b)

Figure 6.2. Progress of the B3LYP/6-31G* geometry optimizations plotted asthe number of iteration versus the energy in kJ/mol relative to the lowest energyfound. (a) Complete course of the optimization where the gaps at iteration 29and 42 represent restarts. High energies are truncated for clarity. (b) Detailedview near the end of the optimization.

The torsional space was explored with DFT calculations on a collection of 16

rotamers. Frequency calculations away from the local energy minima where performed

to gather detailed information of the potential energy surface.

Parameterization. With all of the necessarily X-ray, spectroscopic and QM data in

hand, it was time to fit it all together into a few improved FF parameters. The default

AMBER* FF definition file, amber.fld, was modified with some reasonable guesses

for the parameters which are to be added or modified. We added bond stretch terms for

S–N, where the S is also bound to two N, and for S=O, where the S is also bound to an

N and an O. In other words, the bond stretch terms defining the sulfamide moiety

Page 59: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

59

which where poorly parameterized in the default FF. Parameters defining the

appropriate bond angles, torsions and improper torsions where similarly defined and

given starting values.

Fitting the experimental and calculated data into AMBER* to make an improved, but

still internally consistant, FF was accomplished through the collection of programs and

scripts of Norrby and Liljefors.308 The scripts are partially automated but the user

maintains some control over the procedure through efficient application of either the

simplex or newton-raphson optimizations based on the progress of the convergence.

Results of the parameterization. Starting from the (nearly) optimized QM geometry, a

dihedral angle drive (relaxed potential energy scan) of one of the N–S bonds was

performed both with the default AMBER* FF and with B3LYP/6-31G*. As shown in

Figure 6.3, the torsion energy curve for AMBER* exhibits a roughly negative

correlation to the B3LYP/6-31G*. The minima in the AMBER* are near the maxima

for B3LYP/6-31G*. Figure 6.4 presents the results of the same dihedral angle drive

with the newly parameterized AMBER*. While the fit isn't perfect, all of the features

of the B3LYP/6-31G* curve are reproduced.

Page 60: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

60

Figure 6.3. Drive of one of the S–N torsions using the default AMBER* forcefield in MacroModel 5.5 (solid line) and with B3LYP/6-31G* in Gaussian94(dashed line).

Figure 6.4. Drive of one of the S–N torsions using the reparameterizedAMBER* force field (solid line) and with B3LYP/6-31G* in Gaussian94(dashed line).

Page 61: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

61

Of more practical significance than a reproduction of QM results for a single torsion is

a test of how well the new FF can perform on molecules. Reproduction of the X-ray

structure reported for tetramethylsulfamide model compound (DMAMSO) isn't a

perfect test since the structural determination for this molecule was not of particularly

high quality (RFAC = 0.85) so a comparison to SIKFUN (RFAC = 0.32) was made

instead.

Starting from X-ray coordinates, geometry optimizations of SIKFUN were calculated

using either default AMBER* or the reparameterized version. As summarized in Table

6.1, the new, reparameterized AMBER* parameters were able to maintain the X-ray

geometry of SIKFUN. The ∆E, difference in energies of the minimized and X-ray

geometries, for the new FF were better than achieved with the default parameters but

still not perfect. The new parameters may need further improvement.

Table 6.1: Application of old and new AMBER* to SIKFUN.

AMBER* RMS ∆E

default 0.45 74.8 kJ/mol

reparameterized 0.05 52.6 kJ/mol

Work is in progress to perform more rigorous testing of the new force field parameters

on full-sized inhibitors with the protease active site.

Page 62: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

62

7 CONCLUDING REMARKS

X-ray analysis of a sulfamide derivative of previously studied cyclic urea inhibitors

revealed a unexpected binding mode. The central ring was twisted to flip what would

seem to be the P1' into S2' and the P2' into the S1' pockets of the protease.

Computational studies were initiated to help confirm and understand the nature of this

result. Ab initio calculations were performed to estimate the relative energies of the

symmetric and nonsymmetric conformations. Molecular modelling was used to help

design compounds to test the robustness of the nonsymmetric conformation.

A set of nonsymmetric cyclic sulfamide inhibitors was investigated. These compounds

were designed to be better adapted to the consistently observed nonsymmetric binding

mode. Determination of which side of the inhibitor would adopt the flip turned out to

be nontrivial. CoMFA models were derived for each combination of flips in order to

help guide the SAR. A conventional CoMFA model was subsequently made to aid in

the design of new inhibitors.

Modelling of the cyclic sulfamide inhibitors is impeded by the lack of high quality

empirical force field parameters. Work is in progress to development improved

parameters and preliminary results look promising.

CoMFA models were derived to explain the correlation between structure and binding

data for a set of kinetics data. An extensive and systematic investigation of the

adjustable CoMFA parameters was performed to search for better models. Statistically

improved models were obtained and the practical utility of this procedure is discussed.

Page 63: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

63

8 ACKNOWLEDGEMENTS

I wish to express my sincere gratitude to:

Docent Anders Karlén, my supervisor, for his great patience, accessibility and insight

into complex issues. Especially appreciated was his role in helping me to simplify

my writing when I'd failed to eschew obfuscation.

Professor Anders Hallberg for support throughout this study by providing the excellent

working facilities and through his active encouragement.

All of the chemists who worked so hard to design and synthesize the compounds

investigated in this study. Thanks for giving me something to calculate but

mostly for lending relevance to this study. Special thanks to the prolific

accomplishments of Drs. Johan Hultén and Mathias Alterman.

Dr. Mats Larhed and Docent Uno Svensson for their expert advise both in general

chemistry and diverse topics like auto maintenance and Swedish etymology.

Docent Helena Danielson and her associates for providing the kinetics and affinity

data without which my CoMFA and QSAR models would obviously have been

impossible.

Docent Torsten Unge and associates for solving so many X-ray structures.

Docent Björn Classon and Medivir for the opportunity to work with large datasets.

The PDC at KTH for computational support and facilities. Special thanks to Nils

Smeds who worked so hard on MacroModel when we were the only users.

Dr. Susanne Winiwarter, computational cohort, for her early help with SPL, many

technical discussions, friendship and for being such a good officemate.

Comp Chem students Shane Peterson and Christian Sköld for the promise of

interesting projects together.

Fredrik (Frax) Ax for the friendship and many interesting conversations. Darn shame

you left us for the real world.

Anna Ax née Karlsson for both her valuable friendship and willingness to discuss

ideas regarding synthesis and computational analysis.

Dr. Tero Linnanen for his view from the bench.

Page 64: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

64

Robert Webster for always being my best friend.

Marianne Åström, Gunilla Eriksson and Arne Andersson for their skillful

administrative and technical assistance.

Former advisors: Prof. R. P. Cuila for sparking my interest in organic chemistry; Dr.

Dan Kubose for good laboratory practice; Prof. Anders Liljas for enzymology

and protein crystallography; Prof. Daniel S. Kemp for synthetic organic

chemistry; Prof. W. Todd Wipke for computer programming and computational

chemistry.

Kaisa for dragging me to this winter wonderland but mostly for the love and support.

Я тебя люблю! Daughters Sonia and Ellen for their barely controlled

(im)patience of my work schedule. Thanks to all three for making everything

worthwhile.

Sheila, my mother, for encouraging me to follow my own interests. Siblings Michelle

and Ben for helping me to understand and appreciate children and then for

growing up into real people. Meaw, Popo, Jr, Gail, Carol and cousins for the

sense of family and for the memories of the past.

The Kingdom of Sweden for providing a safe and nurturing environment for my

family, for allowing me to study and work here with essentially the same rights as

a citizen and for the environment of tolerance of my foreign habits and values.

This place could still be improved with California weather and access to better

Salsa but at least I can scratch together everything needed for an authentic

Thanksgiving meal.

This investigation was carried out at the Division of Organic Pharmaceutical

Chemistry, Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala

University, Sweden. Financial support was obtained from the Swedish National Board

for Industrial and Technical Development (NUTEK), the Swedish Foundation for

Strategic Research (SSF) and Medivir AB, Huddinge, Sweden.

Page 65: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

65

9 REFERENCES

(1) Pneumocystis pneumonia--Los Angeles. MMWR Morb. Mortal. Wkly Rep.1981, 30, 250-252.

(2) Kaposi’s sarcoma: the role of HHV-8 and HIV-1 in pathogenesis. http://www-ermm.cbcu.cam.ac.uk/01002733h.htm.

(3) Kaposi's sarcoma and Pneumocystis pneumonia among homosexual men--NewYork City and California. MMWR Morb. Mortal. Wkly Rep. 1981, 30, 305-308.

(4) Update on acquired immune deficiency syndrome (AIDS)--United States.MMWR Morb. Mortal. Wkly Rep. 1982, 31, 507-508, 513-504.

(5) Classification system for human T-lymphotropic virus typeIII/lymphadenopathy-associated virus infections. MMWR Morb. Mortal. WklyRep. 1986, 35, 334-339.

(6) 1993 revised classification system for HIV infection and expanded surveillancecase definition for AIDS among adolescents and adults. MMWR Morb. Mortal.Wkly Rep. 1992, 41, 1-19.

(7) UNAIDS; WHO. AIDS Epidemic Update: December 2001; Joint UnitedNations Programme on HIV/AIDS; World Health Organization: Geneva, 2001.

(8) Gottlieb, M. S.; Schroff, R.; Schanker, H. M.; Weisman, J. D.; Fan, P. T.; Wolf,R. A.; Saxon, A. Pneumocystis carinii pneumonia and mucosal candidiasis inpreviously healthy homosexual men: evidence of a new acquired cellularimmunodeficiency. N. Engl. J. Med. 1981, 305, 1425-1431.

(9) Gallo, R. C.; Salahuddin, S. Z.; Popovic, M.; Shearer, G. M.; Kaplan, M.;Haynes, B. F.; Palker, T. J.; Redfield, R.; Oleske, J.; Safai, B.; et al. Frequentdetection and isolation of cytopathic retroviruses (HTLV-III) from patients withAIDS and at risk for AIDS. Science 1984, 224, 500-503.

(10) Goedert, J. J.; Neuland, C. Y.; Wallen, W. C.; Greene, M. H.; Mann, D. L.;Murray, C.; Strong, D. M.; Fraumeni, J. F., Jr.; Blattner, W. A. Amyl nitritemay alter T lymphocytes in homosexual men. Lancet 1982, 1, 412-416.

(11) Opportunistic infections and Kaposi's sarcoma among Haitians in the UnitedStates. MMWR Morb. Mortal. Wkly Rep. 1982, 31, 353-354, 360-351.

(12) Clumeck, N.; Mascart-Lemone, F.; de Maubeuge, J.; Brenez, D.; Marcelis, L.Acquired immune deficiency syndrome in Black Africans. Lancet 1983, 1, 642.

(13) Pneumocystis carinii pneumonia among persons with hemophilia A. MMWRMorb. Mortal. Wkly Rep. 1982, 31, 365-367.

(14) Unexplained immunodeficiency and opportunistic infections in infants--NewYork, New Jersey, California. MMWR Morb. Mortal. Wkly Rep. 1982, 31, 665-667.

(15) Immunodeficiency among female sexual partners of males with acquiredimmune deficiency syndrome (AIDS) - New York. MMWR Morb. Mortal. WklyRep. 1983, 31, 697-698.

(16) Murphy, T. F. Is AIDS a just punishment? J. Med. Ethics 1988, 14, 154-160.(17) Barre-Sinoussi, F.; Chermann, J. C.; Rey, F.; Nugeyre, M. T.; Chamaret, S.;

Gruest, J.; Dauguet, C.; Axler-Blin, C.; Vezinet-Brun, F.; Rouzioux, C.;Rozenbaum, W.; Montagnier, L. Isolation of a T-Lymphotropic Retrovirus

Page 66: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

66

from a Patient at Risk for Acquired Immune Deficiency Syndrome (AIDS).Science 1983, 220, 868-871.

(18) Popovic, M.; Sarngadharan, M. G.; Read, E.; Gallo, R. C. Detection, isolation,and continuous production of cytopathic retroviruses (HTLV-III) from patientswith AIDS and pre-AIDS. Science 1984, 224, 497-500.

(19) Duesberg, P.; Rasnick, D. The AIDS dilemma: drug diseases blamed on apassenger virus. Genetica 1998, 104, 85-132.

(20) Clavel, F.; Guetard, D.; Brun-Vezinet, F.; Chamaret, S.; Rey, M. A.; Santos-Ferreira, M. O.; Laurent, A. G.; Dauguet, C.; Katlama, C.; Rouzioux, C.; et al.Isolation of a new human retrovirus from West African patients with AIDS.Science 1986, 233, 343-346.

(21) Los Alamos National Laboratory, Los Alamos, NM. Human Retroviruses andAIDS 1999: A Compilation and Analysis of Nucleic Acid and Amino AcidSequences. http://hiv-web.lanl.gov.

(22) Gao, F.; Bailes, E.; Robertson, D. L.; Chen, Y.; Rodenburg, C. M.; Michael, S.F.; Cummins, L. B.; Arthur, L. O.; Peeters, M.; Shaw, G. M.; Sharp, P. M.;Hahn, B. H. Origin of HIV-1 in the chimpanzee Pan troglodytes troglodytes.Nature 1999, 397, 436-441.

(23) Simon, F.; Mauclere, P.; Roques, P.; Loussert-Ajaka, I.; Muller-Trutwin, M. C.;Saragosti, S.; Georges-Courbot, M. C.; Barre-Sinoussi, F.; Brun-Vezinet, F.Identification of a new human immunodeficiency virus type 1 distinct fromgroup M and group O. Nat. Med. 1998, 4, 1032-1037.

(24) Corbet, S.; Muller-Trutwin, M. C.; Versmisse, P.; Delarue, S.; Ayouba, A.;Lewis, J.; Brunak, S.; Martin, P.; Brun-Vezinet, F.; Simon, F.; Barre-Sinoussi,F.; Mauclere, P. env sequences of simian immunodeficiency viruses fromchimpanzees in Cameroon are strongly related to those of humanimmunodeficiency virus group N from the same geographic area. J. Virol.2000, 74, 529-534.

(25) Hahn, B. H.; Shaw, G. M.; De Cock, K. M.; Sharp, P. M. AIDS as a zoonosis:scientific and public health implications. Science 2000, 287, 607-614.

(26) Nahmias, A. J.; Weiss, J.; Yao, X.; Lee, F.; Kodsi, R.; Schanfield, M.;Matthews, T.; Bolognesi, D.; Durack, D.; Motulsky, A.; et al. Evidence forHuman Infection with an HTLV III/LAV-Like Virus in Central Africa, 1959.Lancet 1986, 1, 1279-1280.

(27) Korber, B.; Muldoon, M.; Theiler, J.; Gao, F.; Gupta, R.; Lapedes, A.; Hahn, B.H.; Wolinsky, S.; Bhattacharya, T. Timing the ancestor of the HIV-1 pandemicstrains. Science 2000, 288, 1789-1796.

(28) Chen, Z.; Luckay, A.; Sodora, D. L.; Telfer, P.; Reed, P.; Gettie, A.; Kanu, J.M.; Sadek, R. F.; Yee, J.; Ho, D. D.; Zhang, L.; Marx, P. A. Humanimmunodeficiency virus type 2 (HIV-2) seroprevalence and characterization ofa distinct HIV-2 genetic subtype from the natural range of simianimmunodeficiency virus-infected sooty mangabeys. J. Virol. 1997, 71, 3953-3960.

(29) Haase, A. T. Population biology of HIV-1 infection: viral and CD4+ T celldemographics and dynamics in lymphatic tissues. Annu. Rev. Immunol. 1999, 17, 625-656.

Page 67: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

67

(30) Quinn, T. C. Acute primary HIV infection. JAMA 1997, 278, 58-62.(31) Daar, E. S.; Moudgil, T.; Meyer, R. D.; Ho, D. D. Transient High Levels of

Viremia in Patients with Primary Human Immunodeficiency Virus Type 1Infection. N. Engl. J. Med. 1991, 324, 961-964.

(32) Ratner, L. HIV Life Cycle and Genetic Approaches. Perspect. Drug Dis. Des.1993, 1, 3-22.

(33) Ho, D. D.; Neumann, A. U.; Perelson, A. S.; Chen, W.; Leonard, J. M.;Markowitz, M. Rapid turnover of plasma virions and CD4 lymphocytes in HIV-1 infection. Nature 1995, 373, 123-126.

(34) Wei, X.; Ghosh, S. K.; Taylor, M. E.; Johnson, V. A.; Emini, E. A.; Deutsch,P.; Lifson, J. D.; Bonhoeffer, S.; Nowak, M. A.; Hahn, B. H.; et al. Viraldynamics in human immunodeficiency virus type 1 infection. Nature 1995,373, 117-122.

(35) Piatak, M., Jr.; Saag, M. S.; Yang, L. C.; Clark, S. J.; Kappes, J. C.; Luk, K. C.;Hahn, B. H.; Shaw, G. M.; Lifson, J. D. High levels of HIV-1 in plasma duringall stages of infection determined by competitive PCR. Science 1993, 259,1749-1754.

(36) Arthur, L. O.; Bess, J. W., Jr.; Sowder, R. C., 2nd; Benveniste, R. E.; Mann, D.L.; Chermann, J. C.; Henderson, L. E. Cellular proteins bound toimmunodeficiency viruses: implications for pathogenesis and vaccines. Science1992, 258, 1935-1938.

(37) Turner, B. G.; Summers, M. F. Structural biology of HIV. J. Mol. Biol. 1999,285, 1-32.

(38) Janvier, K.; Petit, C.; Le Rouzic, E.; Schwartz, O.; Benichou, S. HIV auxiliaryproteins: an interface between the virus and the host. AIDS 2000, 14 Suppl. 3,S21-30.

(39) Strebel, K.; Bour, S. Molecular interactions of HIV with host factors. AIDS1999, 13 Suppl. A, S13-24.

(40) Lawn, S. D.; Butera, S. T.; Folks, T. M. Contribution of immune activation tothe pathogenesis and transmission of human immunodeficiency virus type 1infection. Clin. Microbiol. Rev. 2001, 14, 753-777.

(41) National Center for Biotechnology Information. PubMed.http://www.ncbi.nlm.nih.gov:80/entrez/query.fcgi?db=PubMed.

(42) Dalgleish, A. G.; Beverley, P. C.; Clapham, P. R.; Crawford, D. H.; Greaves,M. F.; Weiss, R. A. The CD4 (T4) antigen is an essential component of thereceptor for the AIDS retrovirus. Nature 1984, 312, 763-767.

(43) Gale, L. M.; McColl, S. R. Chemokines: extracellular messengers for alloccasions? Bioessays 1999, 21, 17-28.

(44) Luban, J.; Bossolt, K. L.; Franke, E. K.; Kalpana, G. V.; Goff, S. P. Humanimmunodeficiency virus type 1 Gag protein binds to cyclophilins A and B. Cell1993, 73, 1067-1078.

(45) Luban, J. Absconding with the chaperone: essential cyclophilin-Gag interactionin HIV-1 virions. Cell 1996, 87, 1157-1159.

(46) Whitcomb, J. M.; Hughes, S. H. Retroviral Reverse Transcription andIntegration: Progress and Problems. Annu. Rev. Cell Biol. 1992, 8, 275-306.

Page 68: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

68

(47) Roberts, J. D.; Bebenek, K.; Kunkel, T. A. The accuracy of reversetranscriptase from HIV-1. Science 1988, 242, 1171-1173.

(48) Charneau, P.; Mirambeau, G.; Roux, P.; Paulous, S.; Buc, H.; Clavel, F. HIV-1reverse transcription. A termination step at the center of the genome. J. Mol.Biol. 1994, 241, 651-662.

(49) Zennou, V.; Petit, C.; Guetard, D.; Nerhbass, U.; Montagnier, L.; Charneau, P.HIV-1 genome nuclear import is mediated by a central DNA flap. Cell 2000,101, 173-185.

(50) Yao, X. J.; Subbramanian, R. A.; Rougeau, N.; Boisvert, F.; Bergeron, D.;Cohen, E. A. Mutagenic analysis of human immunodeficiency virus type 1 Vpr:role of a predicted N-terminal alpha-helical structure in Vpr nuclear localizationand virion incorporation. J. Virol. 1995, 69, 7032-7044.

(51) Hazuda, D. J.; Felock, P.; Witmer, M.; Wolfe, A.; Stillmock, K.; Grobler, J. A.;Espeseth, A.; Gabryelski, L.; Schleif, W.; Blau, C.; Miller, M. D. Inhibitors ofstrand transfer that prevent integration and inhibit HIV-1 replication in cells.Science 2000, 287, 646-650.

(52) Gao, K.; Butler, S. L.; Bushman, F. Human immunodeficiency virus type 1integrase: arrangement of protein domains in active cDNA complexes. EMBOJ. 2001, 20, 3565-3576.

(53) Zack, J. A.; Arrigo, S. J.; Weitsman, S. R.; Go, A. S.; Haislip, A.; Chen, I. S.HIV-1 Entry into Quiescent Primary Lymphocytes: Molecular Analysis Revealsa Labile, Latent Viral Structure. Cell 1990, 61, 213-222.

(54) Bukrinsky, M. I.; Stanwick, T. L.; Dempsey, M. P.; Stevenson, M. Quiescent Tlymphocytes as an inducible virus reservoir in HIV-1 infection. Science 1991,254, 423-427.

(55) Marciniak, R. A.; Calnan, B. J.; Frankel, A. D.; Sharp, P. A. HIV-1 Tat proteintrans-activates transcription in vitro. Cell 1990, 63, 791-802.

(56) Jones, K. A.; Peterlin, B. M. Control of RNA initiation and elongation at theHIV-1 promoter. Annu. Rev. Biochem. 1994, 63, 717-743.

(57) Niederman, T. M.; Thielan, B. J.; Ratner, L. Human immunodeficiency virustype 1 negative factor is a transcriptional silencer. Proc. Natl. Acad. Sci. USA1989, 86, 1128-1132.

(58) Emerman, M.; Vazeux, R.; Peden, K. The rev Gene Product of the HumanImmunodeficiency Virus Affects Envelope-Specific RNA Localization. Cell1989, 57, 1155-1165.

(59) Rabson, A. B.; Lin, H. C. NF-kappa B and HIV: linking viral and immuneactivation. Adv. Pharmacol. 2000, 48, 161-207.

(60) Martin-Serrano, J.; Li, K.; Bieniasz, P. D. Cyclin T1 Expression Is Mediated bya Complex and Constitutively Active Promoter and Does Not Limit HumanImmunodeficiency Virus Type 1 Tat Function in Unstimulated PrimaryLymphocytes. J. Virol. 2002, 76, 208-219.

(61) Chen, B. K.; Gandhi, R. T.; Baltimore, D. CD4 down-modulation duringinfection of human T cells with human immunodeficiency virus type 1 involvesindependent activities of vpu, env, and nef. J. Virol. 1996, 70, 6044-6053.

(62) Schubert, U.; Bour, S.; Ferrer-Montiel, A. V.; Montal, M.; Maldarell, F.;Strebel, K. The two biological activities of human immunodeficiency virus type

Page 69: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

69

1 Vpu protein involve two separable structural domains. J. Virol. 1996, 70, 809-819.

(63) Ross, T. M.; Oran, A. E.; Cullen, B. R. Inhibition of HIV-1 progeny virionrelease by cell-surface CD4 is relieved by expression of the viral Nef protein.Curr. Biol. 1999, 9, 613-621.

(64) Jacks, T.; Power, M. D.; Masiarz, F. R.; Luciw, P. A.; Barr, P. J.; Varmus, H. E.Characterization of ribosomal frameshifting in HIV-1 gag-pol expression.Nature 1988, 331, 280-283.

(65) Freed, E. O. HIV-1 gag proteins: diverse functions in the virus life cycle.Virology 1998, 251, 1-15.

(66) Wang, C. T.; Barklis, E. Assembly, processing, and infectivity of humanimmunodeficiency virus type 1 gag mutants. J. Virol. 1993, 67, 4264-4273.

(67) Yuan, X.; Yu, X.; Lee, T. H.; Essex, M. Mutations in the N-terminal region ofhuman immunodeficiency virus type 1 matrix protein block intracellulartransport of the Gag precursor. J. Virol. 1993, 67, 6387-6394.

(68) Walter, P.; Gilmore, R.; Blobel, G. Protein translocation across the endoplasmicreticulum. Cell 1984, 38, 5-8.

(69) Allan, J. S.; Coligan, J. E.; Barin, F.; McLane, M. F.; Sodroski, J. G.; Rosen, C.A.; Haseltine, W. A.; Lee, T. H.; Essex, M. Major glycoprotein antigens thatinduce antibodies in AIDS patients are encoded by HTLV-III. Science 1985,228, 1091-1094.

(70) Matthews, T. J.; Weinhold, K. J.; Lyerly, H. K.; Langlois, A. J.; Wigzell, H.;Bolognesi, D. P. Interaction between the human T-cell lymphotropic virus typeIIIB envelope glycoprotein gp120 and the surface antigen CD4: role ofcarbohydrate in binding and cell fusion. Proc. Natl. Acad. Sci. USA 1987, 84,5424-5428.

(71) Shiraishi, T.; Misumi, S.; Takama, M.; Takahashi, I.; Shoji, S. Myristoylationof human immunodeficiency virus type 1 gag protein is required for efficientenv protein transportation to the surface of cells. Biochem. Biophys. Res.Commun. 2001, 282, 1201-1205.

(72) Garnier, L.; Bowzard, J. B.; Wills, J. W. Recent advances and remainingproblems in HIV assembly. AIDS 1998, 12 Suppl. A, S5-16.

(73) Wilk, T.; Gross, I.; Gowen, B. E.; Rutten, T.; de Haas, F.; Welker, R.;Krausslich, H. G.; Boulanger, P.; Fuller, S. D. Organization of immature humanimmunodeficiency virus type 1. J. Virol. 2001, 75, 759-771.

(74) Ott, D. E.; Coren, L. V.; Chertova, E. N.; Gagliardi, T. D.; Schubert, U.Ubiquitination of HIV-1 and MuLV Gag. Virology 2000, 278, 111-121.

(75) Garrus, J. E.; von Schwedler, U. K.; Pornillos, O. W.; Morham, S. G.; Zavitz,K. H.; Wang, H. E.; Wettstein, D. A.; Stray, K. M.; Cote, M.; Rich, R. L.;Myszka, D. G.; Sundquist, W. I. Tsg101 and the vacuolar protein sortingpathway are essential for hiv-1 budding. Cell 2001, 107, 55-65.

(76) Martin-Serrano, J.; Zang, T.; Bieniasz, P. D. HIV-1 and Ebola virus encodesmall peptide motifs that recruit Tsg101 to sites of particle assembly tofacilitate egress. Nat. Med. 2001, 7, 1313-1319.

(77) Perez, O. D.; Nolan, G. P. Resistance Is Futile. Assimilation of CellularMachinery by HIV-1. Immunity 2001, 15, 687-690.

Page 70: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

70

(78) Demirov, D. G.; Orenstein, J. M.; Freed, E. O. The Late Domain of HumanImmunodeficiency Virus Type 1 p6 Promotes Virus Release in a Cell Type-Dependent Manner. J. Virol. 2002, 76, 105-117.

(79) Fuller, S. D.; Wilk, T.; Gowen, B. E.; Krausslich, H. G.; Vogt, V. M. Cryo-electron microscopy reveals ordered domains in the immature HIV-1 particle.Curr. Biol. 1997, 7, 729-738.

(80) Nermut, M. V.; Hockley, D. J.; Jowett, J. B.; Jones, I. M.; Garreau, M.;Thomas, D. Fullerene-like organization of HIV gag-protein shell in virus-likeparticles produced by recombinant baculovirus. Virology 1994, 198, 288-296.

(81) Kramer, R. A.; Schaber, M. D.; Skalka, A. M.; Ganguly, K.; Wong-Staal, F.;Reddy, E. P. HTLV-III gag protein is processed in yeast cells by the virus pol-protease. Science 1986, 231, 1580-1584.

(82) Kohl, N. E.; Emini, E. A.; Schleif, W. A.; Davis, L. J.; Heimbach, J. C.; Dixon,R. A.; Scolnick, E. M.; Sigal, I. S. Active human immunodeficiency virusprotease is required for viral infectivity. Proc. Natl. Acad. Sci. U. S. A. 1988,85, 4686-4690.

(83) Chaisson, R. E.; Bacchetti, P.; Osmond, D.; Brodie, B.; Sande, M. A.; Moss, A.R. Cocaine use and HIV infection in intravenous drug users in San Francisco.JAMA 1989, 261, 561-565.

(84) Pellegrino, T.; Bayer, B. M. In vivo effects of cocaine on immune cell function.J. Neuroimmunol. 1998, 83, 139-147.

(85) Soderberg, L. S. Immunomodulation by nitrite inhalants may predisposeabusers to AIDS and Kaposi's sarcoma. J. Neuroimmunol. 1998, 83, 157-161.

(86) Heringlake, S.; Ockenga, J.; Tillmann, H. L.; Trautwein, C.; Meissner, D.;Stoll, M.; Hunt, J.; Jou, C.; Solomon, N.; Schmidt, R. E.; Manns, M. P. GBvirus C/hepatitis G virus infection: a favorable prognostic factor in humanimmunodeficiency virus-infected patients? J. Infect. Dis. 1998, 177, 1723-1726.

(87) Tillmann, H. L.; Heiken, H.; Knapik-Botor, A.; Heringlake, S.; Ockenga, J.;Wilber, J. C.; Goergen, B.; Detmer, J.; McMorrow, M.; Stoll, M.; Schmidt, R.E.; Manns, M. P. Infection with GB Virus C and Reduced Mortality amongHIV-Infected Patients. N. Engl. J. Med. 2001, 345, 715-724.

(88) Xiang, J.; Wunschmann, S.; Diekema, D. J.; Klinzman, D.; Patrick, K. D.;George, S. L.; Stapleton, J. T. Effect of Coinfection with GB Virus C onSurvival among Patients with HIV Infection. N. Engl. J. Med. 2001, 345, 707-714.

(89) Mills, S. Back to behavior: prevention priorities in countries with low HIVprevalence. AIDS 2000, 14 Suppl 3, S267-273.

(90) Fichorova, R. N.; Tucker, L. D.; Anderson, D. J. The molecular basis ofnonoxynol-9-induced vaginal inflammation and its possible relevance to humanimmunodeficiency virus type 1 transmission. J. Infect. Dis. 2001, 184, 418-428.

(91) D'Cruz, O. J.; Venkatachalam, T. K.; Uckun, F. M. Structural requirements forpotent human spermicidal activity of dual-function aryl phosphate derivative ofbromo-methoxy zidovudine (compound WHI-07). Biol. Reprod. 2000, 62, 37-44.

(92) Berman, P. W.; Murthy, K. K.; Wrin, T.; Vennari, J. C.; Cobb, E. K.; Eastman,D. J.; Champe, M.; Nakamura, G. R.; Davison, D.; Powell, M. F.; Bussiere, J.;

Page 71: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

71

Francis, D. P.; Matthews, T.; Gregory, T. J.; Obijeski, J. F. Protection of MN-rgp120-immunized chimpanzees from heterologous infection with a primaryisolate of human immunodeficiency virus type 1. J. Infect. Dis. 1996, 173, 52-59.

(93) Jacobson, J. M.; Lowy, I.; Fletcher, C. V.; O'Neill, T. J.; Tran, D. N.; Ketas, T.J.; Trkola, A.; Klotman, M. E.; Maddon, P. J.; Olson, W. C.; Israel, R. J. Single-dose safety, pharmacology, and antiviral activity of the humanimmunodeficiency virus (HIV) type 1 entry inhibitor PRO 542 in HIV-infectedadults. J. Infect. Dis. 2000, 182, 326-329.

(94) Shearer, W. T.; Israel, R. J.; Starr, S.; Fletcher, C. V.; Wara, D.; Rathore, M.;Church, J.; DeVille, J.; Fenton, T.; Graham, B.; Samson, P.; Staprans, S.;McNamara, J.; Moye, J.; Maddon, P. J.; Olson, W. C. Recombinant CD4-IgG2in human immunodeficiency virus type 1-infected children: phase 1/2 study.The Pediatric AIDS Clinical Trials Group Protocol 351 Study Team. J. Infect.Dis. 2000, 182, 1774-1779.

(95) De Clercq, E.; Yamamoto, N.; Pauwels, R.; Balzarini, J.; Witvrouw, M.; DeVreese, K.; Debyser, Z.; Rosenwirth, B.; Peichl, P.; Datema, R.; Thornton, D.;Skerlj, R.; Gaul, F.; Padmanabhan, S.; Bridger, G.; Henson, G.; Abrams, M.Highly potent and selective inhibition of human immunodeficiency virus by thebicyclam derivative JM3100. Antimicrob. Agents Chemother. 1994, 38, 668-674.

(96) Datema, R.; Rabin, L.; Hincenbergs, M.; Moreno, M. B.; Warren, S.; Linquist,V.; Rosenwirth, B.; Seifert, J.; McCune, J. M. Antiviral efficacy in vivo of theanti-human immunodeficiency virus bicyclam SDZ SID 791 (JM 3100), aninhibitor of infectious cell entry. Antimicrob. Agents Chemother. 1996, 40, 750-754.

(97) De Vreese, K.; Reymen, D.; Griffin, P.; Steinkasserer, A.; Werner, G.; Bridger,G. J.; Este, J.; James, W.; Henson, G. W.; Desmyter, J.; Anne, J.; De Clercq, I.The bicyclams, a new class of potent human immunodeficiency virus inhibitors,block viral entry after binding. Antiviral Res. 1996, 29, 209-219.

(98) Este, J. A.; De Vreese, K.; Witvrouw, M.; Schmit, J. C.; Vandamme, A. M.;Anne, J.; Desmyter, J.; Henson, G. W.; Bridger, G.; De Clercq, E. Antiviralactivity of the bicyclam derivative JM3100 against drug-resistant strains ofhuman immunodeficiency virus type 1. Antiviral Res. 1996, 29, 297-307.

(99) Murakami, T.; Yamamoto, N. Roles of chemokines and chemokine receptors inHIV-1 infection. Int. J. Hematol. 2000, 72, 412-417.

(100) Heveker, N. Chemokine receptors as anti-retroviral targets. Curr. Drug Targets2001, 2, 21-39.

(101) Cabrera, C.; Gutierrez, A.; Barretina, J.; Blanco, J.; Litovchick, A.; Lapidot, A.;Clotet, B.; Este, J. A. Anti-HIV activity of a novel aminoglycoside-arginineconjugate. Antiviral Res. 2002, 53, 1-8.

(102) Saha, K.; Zhang, J.; Gupta, A.; Dave, R.; Yimen, M.; Zerhouni, B. Isolation ofprimary HIV-1 that target CD8+ T Lymphocytes using CD8 as a receptor. Nat.Med. 2001, 7, 65-72.

(103) Nagashima, K. A.; Thompson, D. A.; Rosenfield, S. I.; Maddon, P. J.; Dragic,T.; Olson, W. C. Human immunodeficiency virus type 1 entry inhibitors PRO

Page 72: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

72

542 and T-20 are potently synergistic in blocking virus-cell and cell-cell fusion.J. Infect. Dis. 2001, 183, 1121-1125.

(104) Wild, C. T.; Shugars, D. C.; Greenwell, T. K.; McDanal, C. B.; Matthews, T. J.Peptides corresponding to a predictive alpha-helical domain of humanimmunodeficiency virus type 1 gp41 are potent inhibitors of virus infection.Proc. Natl. Acad. Sci. U. S. A. 1994, 91, 9770-9774.

(105) Wild, C.; Oas, T.; McDanal, C.; Bolognesi, D.; Matthews, T. A syntheticpeptide inhibitor of human immunodeficiency virus replication: correlationbetween solution structure and viral inhibition. Proc. Natl. Acad. Sci. U. S. A.1992, 89, 10537-10541.

(106) Root, M. J.; Kay, M. S.; Kim, P. S. Protein design of an HIV-1 entry inhibitor.Science 2001, 291, 884-888.

(107) Li, Q.; Moutiez, M.; Charbonnier, J. B.; Vaudry, K.; Menez, A.; Quemeneur,E.; Dugave, C. Design of a Gag pentapeptide analogue that binds humancyclophilin A more efficiently than the entire capsid protein: new insights forthe development of novel anti-HIV-1 drugs. J. Med. Chem. 2000, 43, 1770-1779.

(108) Braaten, D.; Aberham, C.; Franke, E. K.; Yin, L.; Phares, W.; Luban, J.Cyclosporine A-resistant human immunodeficiency virus type 1 mutantsdemonstrate that Gag encodes the functional target of cyclophilin A. J. Virol.1996, 70, 5170-5176.

(109) Montaner, J. S.; Montessori, V.; Harrigan, R.; O'Shaughnessy, M.; Hogg, R.Antiretroviral therapy: 'the state of the art'. Biomed. Pharmacother. 1999, 53,63-72.

(110) Fischl, M. A.; Richman, D. D.; Grieco, M. H.; Gottlieb, M. S.; Volberding, P.A.; Laskin, O. L.; Leedom, J. M.; Groopman, J. E.; Mildvan, D.; Schooley, R.T.; et al. The efficacy of azidothymidine (AZT) in the treatment of patients withAIDS and AIDS-related complex. A double-blind, placebo-controlled trial. N.Engl. J. Med. 1987, 317, 185-191.

(111) Yarchoan, R.; Mitsuya, H.; Thomas, R. V.; Pluda, J. M.; Hartman, N. R.; Perno,C. F.; Marczyk, K. S.; Allain, J. P.; Johns, D. G.; Broder, S. In vivo activityagainst HIV and favorable toxicity profile of 2',3'-dideoxyinosine. Science1989, 245, 412-415.

(112) Merigan, T. C.; Skowron, G.; Bozzette, S. A.; Richman, D.; Uttamchandani, R.;Fischl, M.; Schooley, R.; Hirsch, M.; Soo, W.; Pettinelli, C.; et al. Circulatingp24 antigen levels and responses to dideoxycytidine in humanimmunodeficiency virus (HIV) infections. A phase I and II study. Ann. Intern.Med. 1989, 110, 189-194.

(113) Riddler, S. A.; Anderson, R. E.; Mellors, J. W. Antiretroviral activity ofstavudine (2',3'-didehydro-3'-deoxythymidine, D4T). Antiviral Res. 1995, 27,189-203.

(114) Soudeyns, H.; Yao, X. I.; Gao, Q.; Belleau, B.; Kraus, J. L.; Nguyen-Ba, N.;Spira, B.; Wainberg, M. A. Anti-human immunodeficiency virus type 1 activityand in vitro toxicity of 2'-deoxy-3'-thiacytidine (BCH-189), a novelheterocyclic nucleoside analog. Antimicrob. Agents Chemother. 1991, 35, 1386-1390.

Page 73: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

73

(115) Daluge, S. M.; Good, S. S.; Faletto, M. B.; Miller, W. H.; St Clair, M. H.;Boone, L. R.; Tisdale, M.; Parry, N. R.; Reardon, J. E.; Dornsife, R. E.; Averett,D. R.; Krenitsky, T. A. 1592U89, a novel carbocyclic nucleoside analog withpotent, selective anti-human immunodeficiency virus activity. Antimicrob.Agents Chemother. 1997, 41, 1082-1093.

(116) Schinazi, R. F.; McMillan, A.; Cannon, D.; Mathis, R.; Lloyd, R. M.; Peck, A.;Sommadossi, J. P.; St Clair, M.; Wilson, J.; Furman, P. A.; Painter, G.; Choi,W.-B.; Liotta, D. C. Selective inhibition of human immunodeficiency virusesby racemates and enantiomers of cis-5-fluoro-1-[2-(hydroxymethyl)-1,3-oxathiolan-5-yl]cytosine. Antimicrob. Agents Chemother. 1992, 36, 2423-2431.

(117) Tisdale, M.; Kemp, S. D.; Parry, N. R.; Larder, B. A. Rapid in vitro selection ofhuman immunodeficiency virus type 1 resistant to 3'-thiacytidine inhibitors dueto a mutation in the YMDD region of reverse transcriptase. Proc. Natl. Acad.Sci. U. S. A. 1993, 90, 5653-5656.

(118) Darque, A.; Valette, G.; Rousseau, F.; Wang, L. H.; Sommadossi, J. P.; Zhou,X. J. Quantitation of intracellular triphosphate of emtricitabine in peripheralblood mononuclear cells from human immunodeficiency virus-infectedpatients. Antimicrob. Agents Chemother. 1999, 43, 2245-2250.

(119) Kim, H. O.; Schinazi, R. F.; Nampalli, S.; Shanmuganathan, K.; Cannon, D. L.;Alves, A. J.; Jeong, L. S.; Beach, J. W.; Chu, C. K. 1,3-dioxolanylpurinenucleosides (2R,4R) and (2R,4S) with selective anti-HIV-1 activity in humanlymphocytes. J. Med. Chem. 1993, 36, 30-37.

(120) Elwell, L. P.; Ferone, R.; Freeman, G. A.; Fyfe, J. A.; Hill, J. A.; Ray, P. H.;Richards, C. A.; Singer, S. C.; Knick, V. B.; Rideout, J. L.; et al. Antibacterialactivity and mechanism of action of 3'-azido-3'-deoxythymidine (BW A509U).Antimicrob. Agents Chemother. 1987, 31, 274-280.

(121) Balzarini, J.; Perno, C. F.; Schols, D.; De Clercq, E. Activity of acyclicnucleoside phosphonate analogues against human immunodeficiency virus inmonocyte/macrophages and peripheral blood lymphocytes. Biochem. Biophys.Res. Commun. 1991, 178, 329-335.

(122) Tan, C. K.; Civil, R.; Mian, A. M.; So, A. G.; Downey, K. M. Inhibition of theRNase H activity of HIV reverse transcriptase by azidothymidylate.Biochemistry 1991, 30, 4831-4835.

(123) Wermuth, G.; Ganellin, C. R.; Lindberg, P.; Mitscher, L. A. Glossary of TermsUsed in Medicinal Chemistry (IUPAC Recommendations 1998). Pure Appl.Chem. 1998, 70, 1129-1143.

(124) Hargrave, K. D.; Proudfoot, J. R.; Grozinger, K. G.; Cullen, E.; Kapadia, S. R.;Patel, U. R.; Fuchs, V. U.; Mauldin, S. C.; Vitous, J.; Behnke, M. L.; Klunder,J. M.; Pal, K.; Skiles, J. W.; McNeil, D. W.; Rose, J. M.; Chow, G. C.; Skoog,M. T.; Wu, J. C.; Schmidt, G.; Engel, W. W.; Eberlein, W. G.; Saboe, T. D.;Campbell, S. J.; Rosenthal, A.; J, A. Novel non-nucleoside inhibitors of HIV-1reverse transcriptase. 1. Tricyclic pyridobenzo- and dipyridodiazepinones. J.Med. Chem. 1991, 34, 2231-2241.

(125) Romero, D. L.; Morge, R. A.; Biles, C.; Berrios-Pena, N.; May, P. D.; Palmer,J. R.; Johnson, P. D.; Smith, H. W.; Busso, M.; Tan, C. K.; et al. Discovery,synthesis, and bioactivity of bis(heteroaryl)piperazines. 1. A novel class of non-

Page 74: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

74

nucleoside HIV-1 reverse transcriptase inhibitors. J. Med. Chem. 1994, 37, 999-1014.

(126) Young, S. D.; Britcher, S. F.; Tran, L. O.; Payne, L. S.; Lumma, W. C.; Lyle, T.A.; Huff, J. R.; Anderson, P. S.; Olsen, D. B.; Carroll, S. S.; et al. L-743, 726(DMP-266): a novel, highly potent nonnucleoside inhibitor of the humanimmunodeficiency virus type 1 reverse transcriptase. Antimicrob. AgentsChemother. 1995, 39, 2602-2605.

(127) Baba, M.; Shigeta, S.; Tanaka, H.; Miyasaka, T.; Ubasawa, M.; Umezu, K.;Walker, R. T.; Pauwels, R.; De Clercq, E. Highly potent and selective inhibitionof HIV-1 replication by 6-phenylthiouracil derivatives. Antiviral Res. 1992, 17,245-264.

(128) Baba, M.; Shigeta, S.; Yuasa, S.; Takashima, H.; Sekiya, K.; Ubasawa, M.;Tanaka, H.; Miyasaka, T.; Walker, R. T.; De Clercq, E. Preclinical evaluationof MKC-442, a highly potent and specific inhibitor of humanimmunodeficiency virus type 1 in vitro. Antimicrob. Agents Chemother. 1994,38, 688-692.

(129) Brennan, T. M.; Taylor, D. L.; Bridges, C. G.; Leyda, J. P.; Tyms, A. S. Theinhibition of human immunodeficiency virus type 1 in vitro by a non-nucleoside reverse transcriptase inhibitor MKC-442, alone and in combinationwith other anti-HIV compounds. Antiviral Res. 1995, 26, 173-187.

(130) Fujiwara, T.; Sato, A.; el-Farrash, M.; Miki, S.; Abe, K.; Isaka, Y.; Kodama,M.; Wu, Y.; Chen, L. B.; Harada, H.; Sugimoto, H.; Hatanaka, M.; Hinuma, Y.S-1153 inhibits replication of known drug-resistant strains of humanimmunodeficiency virus type 1. Antimicrob. Agents Chemother. 1998, 42,1340-1345.

(131) Kashman, Y.; Gustafson, K. R.; Fuller, R. W.; Cardellina, J. H., 2nd;McMahon, J. B.; Currens, M. J.; Buckheit, R. W., Jr.; Hughes, S. H.; Cragg, G.M.; Boyd, M. R. The calanolides, a novel HIV-inhibitory class of coumarinderivatives from the tropical rainforest tree, Calophyllum lanigerum. J. Med.Chem. 1992, 35, 2735-2743.

(132) Corbett, J. W.; Ko, S. S.; Rodgers, J. D.; Jeffrey, S.; Bacheler, L. T.; Klabe, R.M.; Diamond, S.; Lai, C. M.; Rabel, S. R.; Saye, J. A.; Adams, S. P.; Trainor,G. L.; Anderson, P. S.; Erickson-Viitanen, S. K. Expanded-spectrumnonnucleoside reverse transcriptase inhibitors inhibit clinically relevant mutantvariants of human immunodeficiency virus type 1. Antimicrob. AgentsChemother. 1999, 43, 2893-2897.

(133) Cohen, K. A.; Hopkins, J.; Ingraham, R. H.; Pargellis, C.; Wu, J. C.; Palladino,D. E.; Kinkade, P.; Warren, T. C.; Rogers, S.; Adams, J.; al, e. Characterizationof the binding site for nevirapine (BI-RG-587), a nonnucleoside inhibitor ofhuman immunodeficiency virus type-1 reverse transcriptase. J. Biol. Chem.1991, 266, 14670-14674.

(134) Wu, J. C.; Warren, T. C.; Adams, J.; Proudfoot, J.; Skiles, J.; Raghavan, P.;Perry, C.; Potocki, I.; Farina, P. R.; Grob, P. M. A novel dipyridodiazepinoneinhibitor of HIV-1 reverse transcriptase acts through a nonsubstrate bindingsite. Biochemistry 1991, 30, 2022-2026.

Page 75: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

75

(135) Kohlstaedt, L. A.; Wang, J.; Friedman, J. M.; Rice, P. A.; Steitz, T. A. Crystalstructure at 3.5 A resolution of HIV-1 reverse transcriptase complexed with aninhibitor. Science 1992, 256, 1783-1790.

(136) Hazuda, D.; Felock, P.; Hastings, J.; Pramanik, B.; Wolfe, A.; Goodarzi, G.;Vora, A.; Brackmann, K.; Grandgenett, D. Equivalent inhibition of half-site andfull-site retroviral strand transfer reactions by structurally diverse compounds.J. Virol. 1997, 71, 807-811.

(137) Hwang, D. J.; Kim, S. N.; Choi, J. H.; Lee, Y. S. Dicaffeoyl- or digalloylpyrrolidine and furan derivatives as HIV integrase inhibitors. Bioorg. Med.Chem. 2001, 9, 1429-1437.

(138) Jing, N.; Marchand, C.; Guan, Y.; Liu, J.; Pallansch, L.; Lackman-Smith, C.;De Clercq, E.; Pommier, Y. Structure-activity of inhibition of HIV-1 integraseand virus replication by G-quartet oligonucleotides. DNA Cell. Biol. 2001, 20,499-508.

(139) Singh, S. B.; Jayasuriya, H.; Salituro, G. M.; Zink, D. L.; Shafiee, A.;Heimbuch, B.; Silverman, K. C.; Lingham, R. B.; Genilloud, O.; Teran, A.;Vilella, D.; Felock, P.; Hazuda, D. The complestatins as HIV-1 integraseinhibitors. Efficient isolation, structure elucidation, and inhibitory activities ofisocomplestatin, chloropeptin I, new complestatins, A and B, and acid-hydrolysis products of chloropeptin I. J. Nat. Prod. 2001, 64, 874-882.

(140) Myriad Genetics. Press Release: Myriad Genetics Reports Advanced Pre-Clinical Progress with Anti-HIV Drug.http://www.myriad.com/pr/20010920.html.

(141) Senior, K. Budding new HIV therapies? Drug Discov. Today 2001, 6, 1184-1186.

(142) De Clercq, E. New Developments in Anti-HIV Chemotherapy. Curr. Med.Chem. 2001, 8, 1543-1572.

(143) Huff, J. R.; Kahn, J. Discovery and Clinical Development of HIV-1 ProteaseInhibitors. Adv. Protein Chem. 2001, 56, 213-251.

(144) Chen, Z.; Li, Y.; Chen, E.; Hall, D. L.; Darke, P. L.; Culberson, C.; Shafer, J.A.; Kuo, L. C. Crystal Structure at 1.9-Å Resolution of HumanImmunodeficiency Virus (HIV) II Protease Complexed with L-735,524, anOrally Bioavailable Inhibitor of the HIV Proteases. J. Biol. Chem. 1994, 269,26344-26348.

(145) Kempf, D. J.; Marsh, K. C.; Denissen, J. F.; McDonald, E.; Vasavanonda, S.;Flentge, C. A.; Green, B. E.; Fino, L.; Park, C. H.; Kong, X.-P.; Wideburg, N.E.; Saldivar, A.; Ruiz, L.; Kati, W. M.; Sham, H. L.; Robins, T.; Stewart, K. D.;Hsu, A.; Plattner, J. J.; Leonard, J. M.; Norbeck, D. W. ABT-538 is a PotentInhibitor of Human Immunodeficiency Virus Protease and Has High OralBioavailablity in Humans. Proc. Natl. Acad. Sci. USA 1995, 92, 2484-2488.

(146) Krohn, A.; Redshaw, S.; Ritchie, J. C.; Graves, B. J.; Hatada, M. H. NovelBinding Mode of Highly Potent HIV-Protease Inhibitors Incorporating the (R)-Hydroxyethylamine Isostere. J. Med. Chem. 1991, 34, 3340-3342.

(147) Kaldor, S. W.; Kalish, V. J.; Davies, J. F., II; Shetty, B. V.; Fritz, J. E.; Appelt,K.; Burgess, J. A.; Campanale, K. M.; Chirgadze, N. Y.; Clawson, D. K.;Dressman, B. A.; Hatch, S. D.; Khalil, D. A.; Kosa, M. B.; Lubbehusen, P. P.;

Page 76: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

76

Muesing, M. A.; Patick, A. K.; Reich, S. H.; Su, K. S.; Tatlock, J. H. Viracept(Nelfinavir Mesylate, AG1343): A potent, Orally Bioavailable Inhibitor ofHIV-1 Protease. J. Med. Chem. 1997, 40, 3979-3985.

(148) Kim, E. E.; Baker, C. T.; Dwyer, M. D.; Murcko, M. A.; Rao, B. G.; Tung, R.D.; Navia, M. A. Crystal Structure of HIV-1 Protease in Complex with VX-478, a Potent and Orally Bioavailable Inhibitor of the Enzyme. J. Am. Chem.Soc. 1995, 117, 1181-1182.

(149) Sham, H. L.; Kempf, D. J.; Molla, A.; Marsh, K. C.; Kumar, G. N.; Chen, C.M.; Kati, W.; Stewart, K.; Lal, R.; Hsu, A.; Betebenner, D.; Korneyeva, M.;Vasavanonda, S.; McDonald, E.; Saldivar, A.; Wideburg, N.; Chen, X.; Niu, P.;Park, C.; Jayanti, V.; Grabowski, B.; Granneman, G. R.; Sun, E.; Japour, A. J.;Norbeck, D. W.; et al. ABT-378, a highly potent inhibitor of the humanimmunodeficiency virus protease. Antimicrob. Agents Chemother. 1998, 42,3218-3224.

(150) Bold, G.; Fassler, A.; Capraro, H. G.; Cozens, R.; Klimkait, T.; Lazdins, J.;Mestan, J.; Poncioni, B.; Rosel, J.; Stover, D.; Tintelnot-Blomley, M.;Acemoglu, F.; Beck, W.; Boss, E.; Eschbach, M.; Hurlimann, T.; Masso, E.;Roussel, S.; Ucci-Stoll, K.; Wyss, D.; Lang, M. New aza-dipeptide analoguesas potent and orally absorbed HIV-1 protease inhibitors: candidates for clinicaldevelopment. J. Med. Chem. 1998, 41, 3387-3401.

(151) Robinson, B. S.; Riccardi, K. A.; Gong, Y. F.; Guo, Q.; Stock, D. A.; Blair, W.S.; Terry, B. J.; Deminie, C. A.; Djang, F.; Colonno, R. J.; Lin, P. F. BMS-232632, a highly potent human immunodeficiency virus protease inhibitor thatcan be used in combination with other available antiretroviral agents.Antimicrob. Agents Chemother. 2000, 44, 2093-2099.

(152) Poppe, S. M.; Slade, D. E.; Chong, K. T.; Hinshaw, R. R.; Pagano, P. J.;Markowitz, M.; Ho, D. D.; Mo, H.; Gorman, R. R., 3rd; Dueweke, T. J.;Thaisrivongs, S.; Tarpley, W. G. Antiviral activity of the dihydropyrone PNU-140690, a new nonpeptidic human immunodeficiency virus protease inhibitor.Antimicrob. Agents Chemother. 1997, 41, 1058-1063.

(153) Rusconi, S.; La Seta Catamancio, S.; Citterio, P.; Kurtagic, S.; Violin, M.;Balotta, C.; Moroni, M.; Galli, M.; d'Arminio-Monforte, A. Susceptibility toPNU-140690 (Tipranavir) of human immunodeficiency virus type 1 isolatesderived from patients with multidrug resistance to other protease inhibitors.Antimicrob. Agents Chemother. 2000, 44, 1328-1332.

(154) Hodge, C. N.; Aldrich, P. E.; Bacheler, L. T.; Chang, C.-H.; Eyermann, C. J.;Garber, S.; Grubb, M.; Jackson, D. A.; Jadhav, P. K.; Korant, B.; Lam, P. Y. S.;Maurin, M. B.; Meek, J. L.; Otto, M. J.; Rayner, M. M.; Reid, C.; Sharpe, T. R.;Shum, L.; Winslow, D. L.; Erickson-Viitanen, S. Improved Cyclic UreaInhibitors of the HIV-1 Protease: Synthesis, Potency, Resistance Profile,Human Pharmacokinetics and X-Ray Crystal Structure of DMP 450. Chem.Biol. 1996, 3, 301-314.

(155) Phase III trials for new PI. AIDS Patient Care STDS 2001, 15, 174.(156) Gatell, J. M. From amprenavir to GW433908. J. HIV Ther. 2001, 6, 95-99.(157) Zutshi, R.; Franciskovich, J.; Shultz, M.; Schweitzer, B.; Bishop, P.; Wilson,

M.; Chmielewski, J. Targeting the Dimerization Interface of HIV-1 Protease:

Page 77: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

77

Inhibition with Cross-Linked Interfacial Peptides. J. Am. Chem. Soc. 1997, 119,4841-4845.

(158) Zutshi, R.; Chmielewski, J. Targeting the Dimerization Interface forIrreversible Inhibition of HIV-1 Protease. Bioorg. Med. Chem. Lett. 2000, 10,1901-1903.

(159) De Clercq, E. Current lead natural products for the chemotherapy of humanimmunodeficiency virus (HIV) infection. Med. Res. Rev. 2000, 20, 323-349.

(160) Au, T. K.; Lam, T. L.; Ng, T. B.; Fong, W. P.; Wan, D. C. A comparison ofHIV-1 integrase inhibition by aqueous and methanol extracts of Chinesemedicinal herbs. Life Sci. 2001, 68, 1687-1694.

(161) Ng, T. B.; Lam, T. L.; Au, T. K.; Ye, X. Y.; Wan, C. C. Inhibition of humanimmunodeficiency virus type 1 reverse transcriptase, protease and integrase bybovine milk proteins. Life Sci. 2001, 69, 2217-2223.

(162) Creagh, T.; Ruckle, J. L.; Tolbert, D. T.; Giltner, J.; Eiznhamer, D. A.; Dutta,B.; Flavin, M. T.; Xu, Z. Q. Safety and pharmacokinetics of single doses of (+)-calanolide a, a novel, naturally occurring nonnucleoside reverse transcriptaseinhibitor, in healthy, human immunodeficiency virus-negative human subjects.Antimicrob. Agents Chemother. 2001, 45, 1379-1386.

(163) Barnes, J.; Anderson, L. A.; Phillipson, J. D. St John's wort (Hypericumperforatum L.): a review of its chemistry, pharmacology and clinical properties.J. Pharm. Pharmacol. 2001, 53, 583-600.

(164) de Maat, M. M. R.; Hoetelmans, R. M. W.; Mathôt, R. A. A.; van Gorp, E. C.M.; Meenhorst, P. L.; Mulder, J. W.; Beijnen, J. H. Drug interaction between StJohn's wort and nevirapine. AIDS 2001, 15, 420-421.

(165) Piscitelli, S. C.; Gallicano, K. D. Interactions among drugs for HIV andopportunistic infections. N. Engl. J. Med. 2001, 344, 984-996.

(166) Lundgren, J. D.; Phillips, A. N.; Pedersen, C.; Clumeck, N.; Gatell, J. M.;Johnson, A. M.; Ledergerber, B.; Vella, S.; Nielsen, J. O. Comparison of long-term prognosis of patients with AIDS treated and not treated with zidovudine.AIDS in Europe Study Group. JAMA 1994, 271, 1088-1092.

(167) Yeo, J. M. Current and future trials with zidovudine. J. Infect. 1989, 18 Suppl 1,93-96.

(168) Eron, J. J.; Benoit, S. L.; Jemsek, J.; MacArthur, R. D.; Santana, J.; Quinn, J.B.; Kuritzkes, D. R.; Fallon, M. A.; Rubin, M. Treatment with Lamivudine,Zidovudine, or Both in HIV-Positive Patients with 200 to 500 CD4+ Cells perCubic Millimeter. N. Engl. J. Med. 1995, 333, 1662-1669.

(169) Gulick, R. M.; Mellors, J. W.; Havlir, D.; Eron, J. J.; Gonzalez, C.; McMahon,D.; Richman, D. D.; Valentine, F. T.; Jonas, L.; Meibohm, A.; Emini, E. A.;Chodakewitz, J. A.; Deutsch, P.; Holder, D.; Schleif, W. A.; Condra, J. H.Treatment with Indinavir, Zidovudine, and Lamivudine in Adults with HumanImmunodeficiency Virus Infection and Prior Antiretroviral Therapy. N. Engl. J.Med. 1997, 337, 734-739.

(170) Hammer, S. M.; Squires, K. E.; Hughes, M. D.; Grimes, J. M.; Demeter, L. M.;Currier, J. S.; Eron, J. J.; Feinberg, J. E.; Balfour, H. H.; Deyton, L. R.;Chodakewitz, J. A.; Fischl, M. A.; Phair, J. P.; Pedneault, L.; Nguyen, B.-Y.;Cook, J. C.; The AIDS Clinical Trials Group 320 Study Team A Controlled

Page 78: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

78

Trial of Two Nucleoside Analogues plus Indinavir in Persons with HumanImmunodeficiency Virus Infection and CD4 Cell Counts of 200 per CubicMillimeter or Less. N. Engl. J. Med. 1997, 337, 725-733.

(171) Palella, F. J., Jr; Delaney, K. M.; Moorman, A. C.; Loveless, M. O.; Fuhrer, J.;Satten, G. A.; Aschman, D. J.; Holmberg, S. D. Declining Morbidity andMortality Among Patients with Advanced Human Immunodeficiency VirusInfection. N. Engl. J. Med. 1998, 338, 853-860.

(172) Gortmaker, S. L.; Hughes, M.; Cervia, J.; Brady, M.; Johnson, G. M.; Seage, G.R., III; Song, L. Y.; Dankner, W. M.; Oleske, J. M. Effect of CombinationTherapy Including Protease Inhibitors on Mortality among Children andAdolescents Infected with HIV-1. N. Engl. J. Med. 2001, 345, 1522-1528.

(173) Attaran, A.; Sachs, J. Defining and refining international donor support forcombating the AIDS pandemic. Lancet 2001, 357, 57-61.

(174) Steinbrook, R.; Drazen, J. M. AIDS -- Will the Next 20 Years Be Different? N.Engl. J. Med. 2001, 344, 1781-1782.

(175) Herman, J. S.; Easterbrook, P. J. The metabolic toxicities of antiretroviraltherapy. Int. J. STD AIDS 2001, 12, 555-562; quiz 563-554.

(176) Johns, D. R. The other human genome: mitochondrial DNA and disease. Nat.Med. 1996, 2, 1065-1068.

(177) Kakuda, T. N. Pharmacology of nucleoside and nucleotide reverse transcriptaseinhibitor-induced mitochondrial toxicity. Clin. Ther. 2000, 22, 685-708.

(178) Carr, A.; Samaras, K.; Thorisdottir, A.; Kaufmann, G. R.; Chisholm, D. J.;Cooper, D. A. Diagnosis, prediction, and natural course of HIV-1 protease-inhibitor-associated lipodystrophy, hyperlipidaemia, and diabetes mellitus: acohort study. Lancet 1999, 353, 2093-2099.

(179) Carr, A.; Samaras, K.; Chisholm, D. J.; Cooper, D. A. Pathogenesis of HIV-1-protease inhibitor-associated peripheral lipodystrophy, hyperlipidaemia, andinsulin resistance. Lancet 1998, 351, 1881-1883.

(180) Miller, K. D.; Jones, E.; Yanovski, J. A.; Shankar, R.; Feuerstein, I.; Falloon, J.Visceral abdominal-fat accumulation associated with use of indinavir. Lancet1998, 351, 871-875.

(181) Brinkman, K.; Smeitink, J. A.; Romijn, J. A.; Reiss, P. Mitochondrial toxicityinduced by nucleoside-analogue reverse-transcriptase inhibitors is a key factorin the pathogenesis of antiretroviral-therapy-related lipodystrophy. Lancet1999, 354, 1112-1115.

(182) Liang, J. S.; Distler, O.; Cooper, D. A.; Jamil, H.; Deckelbaum, R. J.; Ginsberg,H. N.; Sturley, S. L. HIV protease inhibitors protect apolipoprotein B fromdegradation by the proteasome: A potential mechanism for protease inhibitor-induced hyperlipidemia. Nat. Med. 2001, 7, 1327-1331.

(183) Perelson, A. S.; Essunger, P.; Cao, Y.; Vesanen, M.; Hurley, A.; Saksela, K.;Markowitz, M.; Ho, D. D. Decay characteristics of HIV-1-infectedcompartments during combination therapy. Nature 1997, 387, 188-191.

(184) Zhang, Z.-Q.; Schuler, T.; Zupancic, M.; Wietgrefe, S.; Staskus, K. A.;Reimann, K. A.; Reinhart, T. A.; Rogan, M.; Cavert, W.; Miller, C. J.; Veazey,R. S.; Notermans, D.; Little, S.; Danner, S. A.; Richman, D. D.; Havlir, D.;Wong, J.; Jordan, H. L.; Schacker, T. W.; Racz, P.; Tenner-Racz, K.; Letvin, N.

Page 79: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

79

L.; Wolinsky, S.; Haase, A. T. Sexual Transmission and Propagation of SIVand HIV in Resting and Activated CD4(+) T Cells. Science 1999, 286, 1353-1357.

(185) Kaufmann, G. R.; Cooper, D. A. Antiretroviral therapy of HIV-1 infection:established treatment strategies and new therapeutic options. Curr. Opin.Microbiol. 2000, 3, 508-514.

(186) Wong, J. K.; Gunthard, H. F.; Havlir, D. V.; Zhang, Z. Q.; Haase, A. T.;Ignacio, C. C.; Kwok, S.; Emini, E.; Richman, D. D. Reduction of HIV-1 inblood and lymph nodes following potent antiretroviral therapy and the virologiccorrelates of treatment failure. Proc. Natl. Acad. Sci. U. S. A. 1997, 94, 12574-12579.

(187) Grossman, Z.; Polis, M.; Feinberg, M. B.; Levi, I.; Jankelevich, S.; Yarchoan,R.; Boon, J.; de Wolf, F.; Lange, J. M.; Goudsmit, J.; Dimitrov, D. S.; Paul, W.E. Ongoing HIV dissemination during HAART. Nat. Med. 1999, 5, 1099-1104.

(188) Fraser, C.; Ferguson, N. M.; Anderson, R. M. Quantification of intrinsicresidual viral replication in treated HIV-infected patients. Proc. Natl. Acad. Sci.USA 2001, 98, 15167-15172.

(189) Finzi, D.; Blankson, J.; Siliciano, J. D.; Margolick, J. B.; Chadwick, K.;Pierson, T.; Smith, K.; Lisziewicz, J.; Lori, F.; Flexner, C.; Quinn, T. C.;Chaisson, R. E.; Rosenberg, E.; Walker, B.; Gange, S.; Gallant, J.; Siliciano, R.F. Latent infection of CD4+ T cells provides a mechanism for lifelongpersistence of HIV-1, even in patients on effective combination therapy. Nat.Med. 1999, 5, 512-517.

(190) Heath, S. L.; Tew, J. G.; Szakal, A. K.; Burton, G. F. Follicular dendritic cellsand human immunodeficiency virus infectivity. Nature 1995, 377, 740-744.

(191) Zhang, H.; Dornadula, G.; Beumont, M.; Livornese, L., Jr.; Van Uitert, B.;Henning, K.; Pomerantz, R. J. Human immunodeficiency virus type 1 in thesemen of men receiving highly active antiretroviral therapy. N. Engl. J. Med.1998, 339, 1803-1809.

(192) Chun, T. W.; Davey, R. T., Jr.; Ostrowski, M.; Shawn Justement, J.; Engel, D.;Mullins, J. I.; Fauci, A. S. Relationship between pre-existing viral reservoirsand the re-emergence of plasma viremia after discontinuation of highly activeanti-retroviral therapy. Nat. Med. 2000, 6, 757-761.

(193) García-Lerma, J. G.; Heneine, W. Resistance of human immunodeficiency virustype 1 to reverse transcriptase and protease inhibitors: genotypic andphenotypic testing. J. Clin. Virol. 2001, 21, 197-212.

(194) Rousseau, M. N.; Vergne, L.; Montes, B.; Peeters, M.; Reynes, J.; Delaporte,E.; Segondy, M. Patterns of Resistance Mutations to Antiretroviral Drugs inExtensively Treated HIV-1-Infected Patients With Failure of Highly ActiveAntiretroviral Therapy. J. Acquir. Immune. Defic. Syndr. 2001, 26, 36-43.

(195) Cooper, D. A.; Emery, S. Latent reservoirs of HIV infection: flushing with IL-2? Nat. Med. 1999, 5, 611-612.

(196) Beale, K. K.; Robinson, W. E. Combinations of reverse transcriptase, protease,and integrase inhibitors can be synergistic in vitro against drug-sensitive andRT inhibitor-resistant molecular clones of HIV-1. Antiviral Res. 2000, 46, 223-232.

Page 80: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

80

(197) Kraulis, P. J. MOLSCRIPT: A Program to Produce both Detailed andSchematic Plots of Protein Structures. J. Appl. Crystallography 1991, 24, 946-950.

(198) Merritt, E. A.; Bacon, D. J. Raster3D: Photorealistic Molecular Graphics.Methods Enzymol. 1997, 277, 505-524.

(199) Berman, H. M.; Westbrook, J.; Feng, Z.; Gilliland, G.; Bhat, T. N.; Weissig, H.;Shindyalov, I. N.; Bourne, P. E. The Protein Data Bank. Nucleic Acids Res.2000, 28, 235-242.

(200) Wlodawer, A.; Miller, M.; Jaskolski, M.; Sathyanarayana, B. K.; Baldwin, E.;Weber, I. T.; Selk, L. M.; Clawson, L.; Schneider, J.; Kent, S. B. Conservedfolding in retroviral proteases: crystal structure of a synthetic HIV-1 protease.Science 1989, 245, 616-621.

(201) Bäckbro, K.; Löwgren, S.; Österlund, K.; Atepo, J.; Unge, T.; Hultén, J.;Bonham, N. M.; Schaal, W.; Karlén, A.; Hallberg, A. Unexpected BindingMode of a Cyclic Sulfamide HIV-1 Protease Inhibitor. J. Med. Chem. 1997, 40,898-902.

(202) Toh, H.; Ono, M.; Saigo, K.; Miyata, T. Retrovirla Protease-Like Sequence inthe Yeast Transposon Ty1. Nature 1985, 315, 691-692.

(203) Pearl, L. H.; Taylor, W. R. A structural model for the retroviral proteases.Nature 1987, 329, 351-354.

(204) Navia, M. A.; Fitzgerald, P. M. D.; McKeever, B. M.; Leu, C.-T.; Heimbach, J.C.; Herber, W. K.; Sigal, I. S.; Darke, P. L.; Springer, J. P. Three-DimensionalStructure of Aspartyl Protease from Human Immunodeficiency Virus HIV-1.Nature 1989, 337, 615-620.

(205) Miller, M.; Schneider, J.; Sathyanarayana, B. K.; Toth, M. V.; Marshall, G. R.;Clawson, L.; Selk, L.; Kent, S. B.; Wlodawer, A. Structure of complex ofsynthetic HIV-1 protease with a substrate-based inhibitor at 2.3 A resolution.Science 1989, 246, 1149-1152.

(206) Fitzgerald, P. M.; McKeever, B. M.; VanMiddlesworth, J. F.; Springer, J. P.;Heimbach, J. C.; Leu, C. T.; Herber, W. K.; Dixon, R. A.; Darke, P. L.Crystallographic analysis of a complex between human immunodeficiencyvirus type 1 protease and acetyl-pepstatin at 2.0-A resolution. J. Biol. Chem.1990, 265, 14209-14219.

(207) Friedman, S. H.; DeCamp, D. L.; Sijbesma, R. P.; Srdanov, G.; Wudl, F.;Kenyon, G. L. Inhibition of the HIV-1 protease by fullerene derivatives: modelbuilding studies and experimental verification. J. Am. Chem. Soc. 1993, 115,6506-6509.

(208) Appelt, K. Crystal Structures of HIV-1 Protease-Inhibitor Complexes. Perspect.Drug Dis. Des. 1993, 1, 23-48.

(209) Hosur, M. V.; Bhat, T. N.; Kempf, D. J.; Baldwin, E. T.; Liu, B.; Gulnik, S.;Wideburg, N. E.; Norbeck, D. W.; Appelt, K.; Erickson, J. W. Influence ofStereochemistry on Activity and Binding Modes for C2 Symmetry-Based DiolInhibitors of HIV-1 Protease. J. Am. Chem. Soc. 1994, 116, 847-855.

(210) Schechter, I.; Berger, A. On the Size of the Active Site in Proteases. I. Papain.Biochem. Biophys. Res. Commun. 1967, 27, 157-162.

Page 81: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

81

(211) Silva, A. M.; Cachau, R. E.; Sham, H. L.; Erickson, J. W. Inhibition andCatalytic Mechanism of HIV-1 Aspartic Protease. J. Mol. Biol. 1996, 255, 321-346.

(212) Babine, R. E.; Bender, S. L. Molecular Recognition of Protein-LigandComplexes: Applications to Drug Design. Chem. Rev. 1997, 97, 1359-1472.

(213) Okimoto, N.; Tsukui, T.; Hata, M.; Hoshino, T.; Tsuda, M. HydrolysisMechanism of the Phenylalanine-Proline Peptide Bond Specific to HIV-1Protease: Investigation by the ab Initio Molecular Orbital Method. J. Am.Chem. Soc. 1999, 121, 7349-7354.

(214) Northrop, D. B. Follow the protons: a low-barrier hydrogen bond unifies themechanisms of the aspartic proteases. Acc. Chem. Res. 2001, 34, 790-797.

(215) Clare, M. HIV Protease: Structure-Based Design. Perspect. Drug Dis. Des.1993, 1, 49-68.

(216) van de Waterbeemd, H.; Carter, R. E.; Grassy, G.; Kubinyi, H.; Martin, Y. C.;Tute, M. S.; Willett, P. Glossary of Terms Used in Computational Drug Design(IUPAC Recommendations 1997).; Academic P: San Diego, 1998.

(217) Garrod, C. Twentieth Century Physics; Faculty Publishing: Davis, CA, 1984.(218) Hirst, D. M. A Computational Approach to Chemistry; Blackwell Scientific:

Oxford, 1990.(219) Roothaan, C. C. J. New Developments in Molecular Orbital Theory. Rev.

Modern Phys. 1951, 23, 69-87.(220) Møller, C.; Plesset, M. S. Note on the Approximation treatment for Many-

Electron Systems. Phys. Rev. 1934, 46, 618-622.(221) Foresman, J. B.; Frisch, Æ. Exploring Chemistry Electronic Structure Methods;

Gaussian: Pittsburg, PA, 1996.(222) Hohenberg, P.; Kohn, W. Inhomogeneous Electron Gas. Phys. Rev. 1964,

B864-B871.(223) Kohn, W.; Sham, L. J. Self-Consistent Equations Including Exchange and

Correlation Effects. Phys. Rev. 1965, 140, A1133-A1138.(224) Becke, A. D. Exploring the limits of gradient corrections in density functional

theory. J Comp. Chem. 1999, 20, 63-69.(225) MOPAC 6.0: Quantum Chemistry Program Exchange #455, Indiana University,

1990.(226) Gundertofte, K.; Liljefors, T.; Norrby, P.-O.; Petterson, I. A Comparison of

Conformational Energies Calculated by Several Molecular Mechanics Methods.J. Comp. Chem. 1996, 17, 429-449.

(227) Warshel, A.; Levitt, M. J. Mol. Biol. 1976, 103, 227-249.(228) Bash, P. A.; Field, M. J.; Karplus, M. J. Am. Chem. Soc. 1987, 109, 8092-8094.(229) Cramer, R. D.; Patterson, D. E.; Bunce, J. D. Comparative Molecular-Field

Analysis (Comfa) .1. Effect of Shape On Binding of Steroids to CarrierProteins. J. Am. Chem. Soc. 1988, 110, 5959-5967.

(230) Marshall, G. R.; Barry, C. D.; Bosshard, H. E.; Dammkoehler, R. A.; Dunn, D.A. ; Olson, E. C. and Christofferson, R. E. Ed.; ACS: Washington DC, 1979;Vol. 112, pp 205-226.

(231) Clark, M.; Cramer, R. D. The Probability of Chance Correlation Using PartialLeast- Squares (Pls). Quant. Struct.-Act. Relat. 1993, 12, 137-145.

Page 82: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

82

(232) Hultén, J.; Andersson, H. O.; Schaal, W.; Danielson, H. U.; Classon, B.;Kvarnström, I.; Karlén, A.; Unge, T.; Samuelsson, B.; Hallberg, A. Inhibitors ofthe C2-Symmetric HIV-1 Protease: Nonsymmetric Binding of a SymmetricCylic Sulfamide with Ketoxime Groups in the P2/P2' Side Chains. J. Med.Chem. 1999, 42, 4054-4061.

(233) Schaal, W.; Karlsson, A.; Ahlsén, G.; Lindberg, J.; Andersson, H. O.;Danielson, U. H.; Classon, B.; Unge, T.; Samuelsson, B.; Hultén, J.; Hallberg,A.; Karlén, A. Synthesis and Comparative Molecular Field Analysis (CoMFA)of Symmetric and Nonsymmetric Cyclic Sulfamide HIV-1 Protease Inhibitors.J. Med. Chem. 2001, 44, 155-169.

(234) Alterman, M.; Björsne, M.; Mühlman, A.; Classon, B.; Kvarnström, I.;Danielson, H.; Markgren, P.-O.; Nillroth, U.; Unge, T.; Hallberg, A.;Samuelsson, B. Design and Synthesis of New Potent C2-Symmetric HIV-1Protease Inhibitors. Use of L-Mannaric Acid as a Peptidomimetic Scaffold. J.Med. Chem. 1998, 41, 3782-3792.

(235) Lam, P. Y. S.; Jadhav, P. K.; Eyermann, C. J.; Hodge, C. N.; Ru, Y.; Bacheler,L. T.; Meek, J. L.; Otto, M. J.; Rayner, M. M.; Wong, Y. N.; Chang, C.-H.;Weber, P. C.; Jackson, D. A.; Sharpe, T. R.; Erickson-Viitanen, S. RationalDesign of Potent, Bioavailable, Nonpeptide Cyclic Ureas as HIV ProteaseInhibitors. Science 1994, 263, 380-384.

(236) Sierra, J.; Niño, S.; Volkow, P.; Sereni, D.; Yeni, P.; Staszewski, S.; Gatell, J.;Wang, L.; McMillan, N.; Rousseau, F.; Miralles, G. D. Preliminary profile ofthe antiviral activity, metabolic effects and saftey of DMP-450, a novel cyclicurea protease inhibitor. Antiviral Ther. 2000, 5 suppl.3, 6.

(237) Hultén, J. Cyclic Sulfamides as HIV-1 Protease Inhibitors : Synthesis, X-rayStructure Analysis and Structure-Activity Relationship.; Acta UniversitatisUpsaliensis: Uppsala, 1999.

(238) Hultén, J.; Bonham, N. M.; Nillroth, U.; Hansson, T.; Zuccarello, G.; Bouzide,A.; Åqvist, J.; Classon, B.; Danielsson, U. H.; Karlén, A.; Kvarnström, I.;Samuelsson, B.; Hallberg, A. Cyclic HIV-1 Protease Inhibitors Derived fromMannitol: Synthesis, Inhibitory Potencies, and Computational Predictions ofBinding Affinities. J. Med. Chem. 1997, 40, 885-897.

(239) Nillroth, U.; Vrang, L.; Markgren, P.-O.; Hultén, J.; Hallberg, A.; Danielson, U.H. Human Immunodeficiency Virus Type 1 Proteinase Resistance to SymmetricCyclic Urea Inhibitor Analogs. Antimicrob. Agents Chemother. 1997, 41, 2383-2388.

(240) Smallheer, J. M.; Seitz, S. P. Novel Bicyclic Phosphordiamidate HIV ProteaseInhibitors. Heterocycles 1996, 43, 2367-2376.

(241) Chenera, B.; Desjarlais, R. L.; Finkelstein, J. A.; Eggleston, D. S.; Meek, T. D.;Tomaszek, T. A.; Dreyer, G. B. Nonpeptide Hiv Protease Inhibitors Designedto Replace a Bound Water. Bioorg. Med. Chem. Lett. 1993, 3, 2717-2722.

(242) Kim, C. U.; McGee, L. R.; Krawczyk, S. H.; Harwood, E.; Harada, Y.;Swaminathan, S.; Bischofberger, N.; Chen, M. S.; Cherrington, J. M.; Xiong, S.F.; Giffin, L.; Cundy, K. C.; Lee, A.; Yu, B.; Gulnik, S.; Erickson, J. W. NewSeries of Potent, Orally Bioavailable, Non-Peptidic Cyclic Sulfones as HIV-1 Protease Inhibitors. J. Med. Chem. 1996, 39, 3431-3434.

Page 83: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

83

(243) Jadhav, P. K.; Woerner, F. J. Synthesis of 8-Membered Cyclic Sulfamides:Novel HIV-1 Protease Inhibitors. Tetrahedron Lett. 1995, 36, 6383-6386.

(244) Jadhav, P. K.; Woerner, F. J.; Lam, P. Y.; Hodge, C. N.; Eyermann, C. J.; Man,H. W.; Daneker, W. F.; Bacheler, L. T.; Rayner, M. M.; Meek, J. L.; Erickson-Viitanen, S.; Jackson, D. A.; Calabrese, J. C.; Schadt, M.; Chang, C. H.Nonpeptide cyclic cyanoguanidines as HIV-1 protease inhibitors: synthesis,structure-activity relationships, and X-ray crystal structure studies. J. Med.Chem. 1998, 41, 1446-1455.

(245) Jadhav, P. K.; Man, H. W. Synthesis of 7-membered cyclic oxamides: NovelHIV-1 protease inhibitors. Tetrahedron Lett. 1996, 37, 1153-1156.

(246) Sham, H. L.; Zhao, C.; Stewart, K. D.; Betebenner, D. A.; Lin, S.; Park, C. H.;Kong, X.-P.; Rosenbrook, W., Jr; Herrin, T.; Madigan, D.; Vasavanonda, S.;Lyons, N.; Molla, A.; Saldivar, A.; Mersh, K. C.; McDonald, E.; Wideburg, N.E.; Denissen, J. F.; Robins, T.; Kempf, D. J.; Plattner, D. J.; Norbeck, D. W. ANovel Picomolar Inhibitor of Human Immunodeficiency Virus Type 1 Protease.J. Med. Chem. 1996, 39, 392-397.

(247) Mohamadi, F.; Richards, N. G. J.; Guida, W. C.; Liskamp, R.; Lipton, M.;Caufield, C.; Chang, G.; Hendrickson, T.; Still, W. C. MacroModel -- AnIntegrated Software System for Modeling Organic and Bioorganic MoleculesUsing Molecular Mechanics. J. Comp. Chem. 1990, 11, 440-467.

(248) McDonald, D. Q.; Still, W. C. Amber* Torsional Parameters For the PeptideBackbone. Tetrahedron Lett. 1992, 33, 7743-7746.

(249) Allen, F. H.; Kennard, O. 3D search and research using the CambridgeStructural Database. Chem. Des. Automation News 1993, 8, 31-37.

(250) Lee, C.-H.; Kohn, H. Intra- and Intermolecular α-SulfamidoalkylationReactions. J. Org. Chem. 1990, 55, 6098-6104.

(251) Halgren, T. A. Merck Molecular Force Field .1. Basis, Form, Scope,Parameterization, and Performance of MMFF94. J. Comput. Chem. 1996, 17,490-519.

(252) Alkorta, I. Comparison of Methods to Estimate Geometric and ElectronicProperties on Sulfur Containing Compounds. Theor. Chim. Acta 1994, 89, 1-12.

(253) Mó, O.; dePaz, J. L. G.; Yáñez, M.; Alkorta, I.; Elguero, J.; Goya, P.; Rozas, I.A Molecular Orbital Study of the Conformation (Inversion and RotationalBarriers) and Electronic Properties of Sulfamide. Can. J. Chem. 1989, 67,2227-2236.

(254) Nicholas, J. B.; Vance, R.; Martin, E.; Burke, B. J.; Hopfinger, A. J. AMolecular Mechanics Valence Force Field for Sulfonamides Derived by abInitio Methods. J. Phys. Chem. 1991, 95, 9803-9811.

(255) Becke, A. D. Density-Functional Exchange-Energy Approximation withCorrect Asymptotic Behavior. Phys. Rev. A 1988, 38, 3098–3100.

(256) Lee, C.; Yang, W.; Parr, R. G. Development of the Colle-Salvetti correlation-energy formula into a functional of the electron density. Phys. Rev. B 1988, 37,785-789.

(257) Frisch, M. J.; Trucks, G. W.; Schlegel, H. B.; Gill, P. M. W.; Johnson, B. G.;Robb, M. A.; Cheeseman, J. R.; Keith, T.; Petersson, G. A.; Montgomery, J. A.;Raghavachari, K.; Al-Laham, M. A.; Zakrzewski, V. G.; Ortiz, J. V.; Foresman,

Page 84: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

84

J. B.; Cioslowski, J.; Stefanov, B. B.; Nanayakkara, A.; Challacombe, M.;Peng, C. Y.; Ayala, P. Y.; Chen, W.; Wong, M. W.; Andres, J. L.; Replogle, E.S.; Gomperts, R.; Martin, R. L.; Fox, D. J.; Binkley, J. S.; Defrees, D. J.; Baker,J.; Stewart, J. P.; Head-Gordon, M.; Gonzalez, C.; Pople, J. A. Gaussian 94(Revision D.4); Gaussian, Inc.: Pittsburgh PA, 1995.

(258) Han, Q.; Chang, C. H.; Li, R.; Ru, Y.; Jadhav, P. K.; Lam, P. Y. Cyclic HIVProtease Inhibitors: Design and Synthesis of Orally Bioavailable, PyrazoleP2/P2' Cyclic Ureas with Improved Potency. J. Med. Chem. 1998, 41, 2019-2028.

(259) Still, W. C.; Tempczyk, A.; Hawley, R. C.; Hendrickson, T. SemianalyticalTreatment of Solvation For Molecular Mechanics and Dynamics. J. Am. Chem.Soc. 1990, 112, 6127-6129.

(260) Wlodawer, A.; Erickson, J. W. Structure-Based Inhibitors of HIV-1 Protease.Annu. Rev. Biochem. 1993, 62, 543-585.

(261) Garg, R.; Gupta, S. P.; Gao, H.; Babu, M. S.; Debnath, A. K.; Hanch, C.Comparative Quantitative Structure-Activity Relationship Studies on Anti-HIVDrugs. Chem. Rev. 1999, 99, 3525-3601.

(262) Waller, C. L.; Oprea, T. I.; Giolitti, A.; Marshall, G. R. Three-DimensionalQSAR of Human Immunodeficiency Virus (I) Protease Inhibitors. 1. A CoMFAStudy Employing Eperimentally-Determined Alignment Rules. J. Med. Chem.1993, 36, 4152-4160.

(263) Oprea, T. I.; Waller, C. L.; Marshall, G. R. Three-Dimensional QuantitativeStructure-Activity Relationaship of Human Immunodeficiency Virus (I)Protease Inhibitors. 2. Predictive Power Using Limited Exploration of AlternateBinding Modes. J. Med. Chem. 1994, 37, 2206-2215.

(264) Oprea, T. I.; Waller, C. L.; Marshall, G. R. 3D-QSAR of HumanImmunodeficiency Virus (I) Protease Inhibitors. III. Interpretation of CoMFAResults. Drug Des. Discov. 1994, 12, 29-51.

(265) Kroemer, R. T.; Ettmayer, P.; Hecht, P. 3D-Quantitative Structure-ActivityRelationships of Human Immunodeficiency Virus Type-1 Proteinase Inhibitors:Comparative Molecular Field Analysis of 2-Heterosubstituted StatineDerivatives--Implications for the Design of Novel Inhibitors. J. Med. Chem.1995, 38, 4917-4928.

(266) Debnath, A. K. Comparative molecular field analysis (CoMFA) of a series ofsymmetrical bis-benzamide cyclic urea derivatives as HIV-1 proteaseinhibitors. J. Chem. Inf. Comput. Sci. 1998, 38, 761-767.

(267) Debnath, A. K. Three-Dimensional Quantitative Structure-ActivityRelationship Study on Cyclic Urea Derivatives as HIV-1 Protease Inhibitors:Application of Comparative Molecular Field Analysis. J. Med. Chem. 1999, 42,249-259.

(268) Kulkarni, S. S.; Kulkarni, V. M. Structure Based Predictions of BindingAffinity of Human Immunodeficiency Virus-1 Protease Inhibitors. J. Chem. Inf.Comput. Sci. 1999, 39, 1128-1140.

(269) Jayatilleke, P. R.; Nair, A. C.; Zauhar, R.; Welsh, W. J. Computational Studieson HIV-1 Protease Inhibitors: Influence of Calculated Inhibitor-Enzyme

Page 85: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

85

Binding Affinities on the Statistical Quality of 3D-QSAR CoMFA Models. J.Med. Chem. 2000, 43, 4446-4451.

(270) Ortiz, A. R.; Pisabarro, M. T.; Gago, F.; Wade, R. C. Prediction of DrugBinding Affinities by Comparative Binding Energy Analysis. J. Med. Chem.1995, 38, 2681-2691.

(271) Silverman, B. D.; Platt, D. E. Comparative Molecular Moment Analysis(CoMMA): 3D-QSAR without Molecular Superposition. J. Med. Chem. 1996,39, 2129-2140.

(272) Hopfinger, A. J.; Wang, S.; Tokarski, J. S.; Jin, B.; Alburquerque, M. G.;Madhav, P. J.; Duraiswami, C. Construction of 3D-QSAR models using the 4D-QSAR analysis formalism. J. Am. Chem. Soc. 1997, 119, 10509-10524.

(273) Hämäläinen, M. D.; Markgren, P.-O.; Schaal, W.; Karlén, A.; Classon, B.;Vrang, L.; Samuelsson, B.; Hallberg, A.; Danielson, U. H. Characterization of aSet of HIV-1 Protease Inhibitors Using Binding Kinetics Data from aBiosensor-Based Screen. J. Biomol. Screen. 2000, 5, 353-360.

(274) Schaal, W.; Markgren, P.-O.; Hämäläinen, M. D.; Danielson, U. H.; Hallberg,A.; Samuelsson, B.; Karlén, A. Comparative Molecular Field Analysis(CoMFA) of the Association and Dissociation Rate Constants of a Diverse Setof HIV-1 Protease Inhibitors. Manuscript in preparation.

(275) Guttendorf, R. J. The Emerging Role of A.D.M.E. in Optimizing DrugDiscovery and Design. Network Sci. 1996, February,http://www.netsci.org/Science/Special/feature06.html.

(276) Kumar, G. N.; Surapaneni, S. Role of drug metabolism in drug discovery anddevelopment. Med. Res. Rev. 2001, 21, 397-411.

(277) Devlin The Impact of High Throughput Organic Synthesis on R&D in Bio-Based Industries. Network Sci. 1996, March,http://www.netsci.org/Science/Combichem/feature14.html.

(278) Otto, A. Excitation of Nonradiative Surface Plasma Waves in Silver by theMethod of Frustrated Total Reflection. Z. Phys. 1968, 216, 398-410.

(279) Nice, E. C.; Catimel, B. Instrumental biosensors: new perspectives for theanalysis of biomolecular interactions. Bioessays 1999, 21, 339-352.

(280) Mullett, W. M.; Lai, E. P.; Yeung, J. M. Surface plasmon resonance-basedimmunoassays. Methods 2000, 22, 77-91.

(281) Myszka, D. G.; Rich, R. L. Implementing surface plasmon resonancebiosensors in drug discovery. Pharm. Sci. Technol. Today 2000, 3, 310-317.

(282) Rich, R. L.; Myszka, D. G. Advances in surface plasmon resonance biosensoranalysis. Curr. Opin. Biotechnol. 2000, 11, 54-61.

(283) Quinn, J. G.; O'Kennedy, R. Biosensor-based estimation of kinetic andequilibrium constants. Anal Biochem 2001, 290, 36-46.

(284) Markgren, P.-O.; Hämäläinen, M.; Danielson, U. H. Screening of CompoundsInteracting with HIV-1 Proteinase Using Optical Biosensor Technology. Analyt.Bioch. 1998, 265, 340-350.

(285) Markgren, P. O.; Hamalainen, M.; Danielson, U. H. Kinetic analysis of theinteraction between HIV-1 protease and inhibitors using optical biosensortechnology. Anal. Biochem. 2000, 279, 71-78.

Page 86: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

86

(286) Alterman, M.; Sjöbom, H.; Säfsten, P.; Markgren, P. O.; Danielson, U. H.;Hämäläinen, M.; Löfås, S.; Hultén, J.; Classon, B.; Samuelsson, B.; Hallberg,A. P1/P1' modified HIV protease inhibitors as tools in two new sensitivesurface plasmon resonance biosensor screening assays. Eur. J. Pharm. Sci.2001, 13, 203-212.

(287) Markgren, P.-O.; Lindgren, M. T.; Gertow, K.; Karlsson, R.; Hämäläinen, M.;Danielson, U. H. Determination of interaction kinetic constants for HIV-1protease inhibitors using optical biosensor technology. Anal. Biochem. 2001,291, 207-218.

(288) Porter, D. J. T.; Hanlon, M. H.; Carter, L. H., 3rd; Danger, D. P.; Furfine, E. S.Effectors of HIV-1 Protease Peptidolytic Activity. Biochemistry 2001, 40,11131-11139.

(289) Wall, L.; Christiansen, T.; Schwartz, R. L. Programming Perl; O'Reilly &Associates, Inc: Sebastapol, CA, 1996.

(290) Maschera, B.; Darby, G.; Palu, G.; Wright, L. L.; Tisdale, M.; Myers, R.; Blair,E. D.; Furfine, E. S. Human Immunodeficiency Virus. Mutations in the ViralProtease that Confer Resistance to Saquinavir Increase the Dissociation RateConstant of the Protease-Saquinavir Complex. J. Biol. Chem. 1996, 271, 33231-33235.

(291) Markland, W.; Rao, B. G.; Parsons, J. D.; Black, J.; Zuchowski, L.; Tisdale, M.;Tung, R. Structural and Kinetic Analyses of the Protease from an Amprenavir-Resistant Human Immunodeficiency Virus Type 1 Mutant Rendered Resistantto Saquinavir and Resensitized to Amprenavir. J. Virol. 2000, 74, 7636-7641.

(292) Nascimbeni, M.; Lamotte, C.; Peytavin, G.; Farinotti, R.; Clavel, F. Kinetics ofantiviral activity and intracellular pharmacokinetics of humanimmunodeficiency virus type 1 protease inhibitors in tissue culture. Antimicrob.Agents Chemother. 1999, 43, 2629-2634.

(293) Markgren, P.-O.; Schaal, W.; Hämäläinen, M. D.; Karlén, A.; Hallberg, A.;Samuelsson, B.; Danielson, U. H. Structure-interaction kinetic relationships forHIV-1 protease inhibitors. Manuscript in preparation.

(294) Maren, T. H.; Clare, B. W.; Supuran, C. T. Structure-Activity Studies ofSulfonamide Carbonic Anhydrase Inhibitors. Roumanian Chem. Quart. Rev.1994, 2, 259-282.

(295) Clare, B. W.; Supuran, C. T. Carbonic anhydrase inhibitors. Part 41.Quantitative Structure-Activity Correlations Involving Kinetic Rate Constantsof 20 Sulfonamide Inhibitors from a Non-Congeneric Series. Eur. J. Med.Chem. 1997, 32, 311-319.

(296) Andersson, K.; Choulier, L.; Hamalainen, M. D.; van Regenmortel, M. H.;Altschuh, D.; Malmqvist, M. Predicting the Kinetics of Peptide-AntibodyInteractions Using a Multivariate Experimental Design of Sequence andChemical Space. J. Mol. Recognit. 2001, 14, 62-71.

(297) Sybyl 6.7: Tripos Inc., 1699 South Hanley Rd., St. Louis, MO 63144, USA.(298) Bursi, R.; Grootenhuis, P. D. J. Comparative Molecular Field Analysis and

Energy Interaction Studies of Thrombin-Inhibitor Complexes. J. Comput.-AidedMol. Des. 1999, 13, 221-232.

Page 87: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

87

(299) Dinan, L.; Hormann, R. E.; Fujimoto, T. An Extensive Ecdysteroid CoMFA. J.Comput. Aided Mol. Des. 1999, 13, 185-207.

(300) Cho, S. J.; Tropsha, A. Cross-Validated R2-Guided Region Selection forComparative Molecular Field Analysis: A Simple Method to AchieveConsistent Results. J. Med. Chem. 1995, 38, 1060-1066.

(301) Wold, S. Validation of QSARs. Quant. Struct.-Act. Relat. 1991, 10, 191-193.(302) Bush, B. L.; Nachbar, R. B., Jr Sample-Distance Partial Least Squares: PLS

Optimized for Many Variables, with Application to CoMFA. J. Comput.-AidedMol. Des. 1993, 7, 587-619.

(303) Hodge, C. N.; Lam, P. Y. S.; Eyermann, C. J.; Jadhav, P. K.; Ru, Y.;Fernandez, C. H.; De Lucca, G. V.; Chang, C.-H.; Kaltenbach, R. F., III;Holler, E. R.; Woerner, F.; Daneker, W. F.; Emmett, G.; Calabrese, J. C.;Aldrich, P. E. Calculated and Experimental Low-Energy Conformations ofCyclic Urea HIV Protease Inhibitors. J. Am. Chem. Soc. 1998, 120, 4570-4581.

(304) Pérez, C.; Pastor, M.; Ortiz, A. R.; Gago, F. Comparative Binding EnergyAnalysis of HIV-1 Protease Inhibitors: Incorporation of Solvent Effects andValidation as a Powerful Tool in Receptor-Based Drug Design. J. Med. Chem.1998, 41, 836-852.

(305) Hunger, J.; Huttner, G. Optimization and analysis of force field parameters bycombination of genetic algorithms and neural networks. J. Comp. Chem. 1999,20, 455-471.

(306) Faller, R.; Schmitz, H.; Biermann, O.; Müller-Plathe, F. Automaticparameterization of force fields for liquids by simplex optimization. J Comp.Chem. 1999, 20, 1009-1017.

(307) Wang, J.; Kollman, P. A. Automatic Parameterization of Force Field bySystematic Search and Genetic Algorithms. J. Comp. Chem. 2001, 22, 1219-1228.

(308) Norrby, P.-A.; Liljefors, T. Automated Molecular Mechanics Parameterizationwith Simultaneous Utilization of Experimental and Quantum Mechanical Data.J. Comp. Chem. 1998, 19, 1146-1166.

(309) Maple, J. R.; Hwang, M.-J.; Stockfisch, T. P.; Dinur, U.; Waldman, M.; Ewig,C. S.; Hagler, A. T. Derivation of Class II Force Fields. I. Methodology andQuantumForce Field for the Alkyl Functional Group and Alkane Molecules. J.Comp. Chem. 1994, 15, 162-182.

(310) Chakravorty, S.; Reynolds, C. H. Improved AMBER* torsional parameters forthe N-N rotational barrier in diacylhydrazines. J. Mol. Graph. Model. 1999, 17,315-324.

(311) Rodellas, C.; Martinez-Ripoll, M.; Garcia-Blanco, S. 2-Phenyl-3-oxo-5,6-dimethyl-1,2,6-thiadiazine-1,1-dioxide analgesic and antiinflammatory agent.Acta Cryst. 1984, A40, 64-.

(312) Elguero, J.; Goya, P.; Nieves, R.; Ochoa, C.; Rodellas, C.; Martinez-Ripoll, M.;Garcia-Blanco, S. Proton and C-13 Nmr and Crystallographic Study ofSubstituted 1,2,6-Thiadiazinones - Comparison With Related Pyrazoles. J.Chem. Res. 1988, 94-95.

Page 88: Computational Studies of HIV-1 Protease Inhibitors161222/FULLTEXT01.pdfComputational Studies of HIV-1 Protease Inhibitors. Acta Universitatis Upsaliensis. Comprehensive Summaries of

88

(313) Jordan, T.; Smith, H. W.; Lohr, L. L., Jr; Lipscomb, W. N. X-Ray StructureDetermination of (CH3)2NSO2N(CH3)2 and LCAO-MO Study of MultipleBonding in Sulfones. J. Am. Chem. Soc. 1963, 85, 846-851.

(314) Goya, P.; Nieves, R.; Ochoa, C.; Rodellas, C.; Martinez-Ripoll, M.; Garcia-Blanco, S. 1,2,6-Thiadiazin-3,5(2h,6h)-Dione 1,1-Dioxide Derivatives -Crystal-Structure, Physicochemical and Biological Properties. Can. J. Chem.1987, 65, 298-302.

(315) Koch, P.; Boelsterli, J. J.; Hirst, D. R.; Walkinshaw, M. D. A Novel MetabolicPathway For Benzothiadiazoles - X-Ray Molecular-Structure of 5-Chloro-4-(4,5-Dihydroimidazol-2- Ylamino)-1,3-Dimethyl-1,3-Dihydro-2,1,3-Benzothiadiazole 2,2- Dioxide. J. Chem. Soc. Perkin Trans. 2 1990, 1705-1708.

(316) Street, J. P. Structure of the biimidazole dimer obtained from a bridged N,N'-diimidazolyl sulfone. Acta Cryst. 1991, C47, 411-414.

(317) Török, F.; Páldi, E.; Dobos, S.; Fogarasi, G. On the IR and NMR Spectra of the[(CH3)2N]2S, [(CH3)2N]2SO, [(CH3)2N]2SO2 and (CH3)2NSO2ClMolecules. Acta Chim. Acad. Sci. Hungaricae 1970, 63, 417-423.

(318) Brunvoll, J.; Hargittai, I. On the Correlation of Geometric and VibrationalParameters of the SO2 Groups in Sulfone Molecules. Acta Chim. Acad. Sci.Hungaricea 1978, 96, 337-354.

(319) Remizov, A. B.; Butenko, G. G. [Polarized IR Spectra of CrystallineRSO2N(CH3)2 Compounds and Molecular Conformations]. Zh. Strukt. Khim.1979, 20, 63-70.


Recommended